EP3500588A1 - Procédé de préparation de formulations liquides hautement concentrées contenant des biomolécules - Google Patents

Procédé de préparation de formulations liquides hautement concentrées contenant des biomolécules

Info

Publication number
EP3500588A1
EP3500588A1 EP17754327.9A EP17754327A EP3500588A1 EP 3500588 A1 EP3500588 A1 EP 3500588A1 EP 17754327 A EP17754327 A EP 17754327A EP 3500588 A1 EP3500588 A1 EP 3500588A1
Authority
EP
European Patent Office
Prior art keywords
salts
acid
protein
process according
biomolecule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP17754327.9A
Other languages
German (de)
English (en)
Inventor
Patrick Garidel
Sven BAHRENBURG
Torsten Schultz-Fademrecht
Andrea EIPERLE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim International GmbH
Original Assignee
Boehringer Ingelheim International GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim International GmbH filed Critical Boehringer Ingelheim International GmbH
Publication of EP3500588A1 publication Critical patent/EP3500588A1/fr
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/34Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation

Definitions

  • the invention relates to an improved process for the preparation of highly concentrated liquid formulations containing biomolecules.
  • Biomolecule stability, ionic strength, pH value, and biomolecule concentration as well as biomolecule integrity are among the principal parameters to be controlled.
  • Roberts and colleagues have studied the effects of specific ions (sodium chloride, calcium chloride, sodium sulfate, and sodium thiocyanate) and buffers (acetate, citrate, phosphate, histidine, succinate and tris) on protein-protein interactions in a monoclonal antibody (Roberts D., Keeling R., Tracka M., van der Walle C.F., Uddin S., Warwicker J., Curtis R. (2015), Specific ion and buffer effects on protein-protein interactions of a monoclonal antibody, Molecular Pharmaceutics 2015, 12, 179-193).
  • specific ions sodium chloride, calcium chloride, sodium sulfate, and sodium thiocyanate
  • buffers acetate, citrate, phosphate, histidine, succinate and tris
  • a protein solution may have to be modified several times to facilitate unit operations, storage, and/or formulation.
  • Each stage is likely to involve solution exchanges using processes that broadly qualify as filtration - ultrafiltration (UF), size exclusion chromatography (SEC), diafiltration (DF), and counter-current dialysis - alone or in combination.
  • UF ultrafiltration
  • SEC size exclusion chromatography
  • DF diafiltration
  • counter-current dialysis - alone or in combination These methods help to condition the protein and alter solution conditions to specified ranges (Janson H.-C. (ed.). (201 1 ), Protein Purification, 3 rd edition, Wiley, New Jersey).
  • UF/DF ultrafiltration/diafiltration
  • Brose D.J., Dosmar M., Jornitz M.W. (2002), Membrane filtration, Pharmaceutical biotechnology, 14, pp. 213-279 ultrafiltration/diafiltration
  • UF/DF is used extensively in downstream processing to concentrate proteins, exchange buffer solutions, condition proteins for such downstream processes as chromatography, and recover the protein in the concentration and buffer solution required for formulation
  • Marshak D.R. Kadonaga J.T., Burgess R.R., Knuth M.W., Brennman W.A., Lin S.-H. (1996), Protein Purification and Characterisation, Cold Spring Harbor Laboratory Press.
  • Ultrafiltration / diafiltration is the method usually employed to adjust the pH value, alter the solution's ionic profile/excipient composition, and/or attain target protein concentrations.
  • UF/DF is usually performed in tangential flow filtration (hereinafter also abbreviated as "TFF") mode, which is also called cross-flow, where feed-solution flow runs parallel to the membrane and thus perpendicular to the filtrate flow.
  • TFF tangential flow filtration
  • This setup sweeps retained molecules along the membrane surface, out of the membrane chamber, and back to the retentate vessel - offering significantly higher process throughput than dead-end operations (Flickinger, M. C. (ed.) (2013), Downstream industrial biotechnology, John Wiley & Sons, Hoboken Ney Jersey).
  • TFF often produces concentration polarization, formation of a high concentration gradient and a boundary layer of highly concentrated solutes at the membrane's upstream surface.
  • protein adsorption, denaturation, aggregation, or precipitation may foul the membrane (Field R. (2010), Fundamentals of fouling, in: Peinemann K.-V., Pereira Nunes S., Membranes for water treatment, Volume 4, Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, chapter 1 , 1 -23).
  • the performance of these systems depends almost entirely on the rate at which retained solutes are transported away from the membrane and back into the bulk solution (Bowen W.R., Jenner R. (1995), Theoretical descriptions of membrane filtration of colloids and fine particles: an assessment and review, Adv. Colloid Interface Sci 56, 141 -200). This phenomenon is known as concentration polarization.
  • the diafiltration is a technique that uses permeable or porous membrane filters to remove, replace, or lower the concentration of salts or solvents from solutions containing biomolecules.
  • the process uses the membrane filters to separate the components of solutions and suspensions based on their molecular size.
  • the solution retained by the membrane is known as the concentrate or retentate.
  • the solution that passes through the membrane is known as the filtrate or permeate.
  • feedstock is repeatedly circulated over the membrane and returned to the retentate vessel, where new liquid medium such as a buffer is added, while permeate removed.
  • the diafiltration medium corresponds to the feed medium added to the system, whereas the permeate is the filtration medium removed from the system.
  • Figure 1 B a schematic representation of an ultrafiltration (UF) step is illustrated. Ultrafiltration per se follows the same concept and is based on the same schematic setup as shown for the diafiltration in Figure 1A, but without the addition of new liquid medium.
  • CA 2 643 508 A1 a process is described for obtaining a human albumin solution, with a high capacity for binding molecules which comprises:
  • a combination of diafiltration steps for example, used to detoxify albumin in the blood or plasma of a patient, with the aim to remove substances that are bound to albumin.
  • the albumin is heated, i.e. a step of virus inactivation by pasteurisation is performed, with the albumin stabilised in the presence of at least one amino acid and sodium chloride.
  • This process allows removing compounds bound to the albumin such as lipids, fatty acids, because these compounds reduce the binding capacity of albumin.
  • a heating of the solution between a first and a second diafiltration is not performed in the present invention.
  • WO 91/00290 A1 relates to a method for purifying a protein from multivalent metal ions bound thereto, these ions being released from the protein by exchanging the ions with monovalent metal ions, whereafter the multivalent metal ions are removed.
  • the release and removal of these ions are effected, in particular by methods of diafiltration or gel filtration.
  • cleansing proteins such as albumin and gammaglobulin from multivalent metal ions, e.g. aluminium, iron or lead, which are bound to the proteins, it is used a combination of two gel-filtration steps including water and salt solutions up to 1 M, in order to remove the multivalent metal ions and exchanging them against sodium, potassium or ammonium ions.
  • WO 2002/051979 A2 it is provided a method to remove citrate, aluminium, multivalent ions and contaminants from proteins by adjusting the pH of a solution containing the protein to a pH from about 7 to about 10, diafiltering the aqueous solution against pure water to thereby provide a filtrate comprising the multivalent ions and a retentate comprising the protein.
  • WO 02/096457 A2 is directed to stable liquid formulations of antibodies suitable for parenteral administration.
  • aqueous solutions which have high concentrations of therapeutical antibodies which may be used to produce therapeutical liquid formulations, uses, such as medical uses, of the stable liquid formulations and processes for the production of the stable liquid formulations.
  • a process for the preparation of a therapeutical liquid formulation comprises an antibody at a concentration of more than 50 mg/ml, wherein in a first step an antibody solution in a suitable buffer is concentrated to a concentration in the range from about 10 mg/ml to about 50 mg/ml; in a second step, the concentrated solution obtained in the first step is diafiltered with an aqueous solution of at least one acidic component, optionally containing MgC and/or CaC ⁇ and/or further suitable additives; and, in a third step, the solution obtained in the second step is further concentrated to a concentration of more than 50 mg/ml. Therefore, the process uses the sequence of a concentration step / a diafiltration step / a concentration step.
  • the concentration step may be performed with an ultrafiltration system.
  • WO 2004/042012 A2 it is also presented a process to concentrate macromolecules. It is provided a method for concentrating a macromolecule from an aqueous starting solution having solution components, the solution components comprising the macromolecule and an organic polymer, the method comprising: (1 ) subjecting the aqueous starting solution to ultrafiltration to concentrate the macromolecule such that a first retentate solution is produced, (2) adjusting the conductivity of the first retentate solution such that precipitation of the solution components induced by the organic polymer is substantially prevented or substantially reversed to produce a second retentate solution, and
  • the conductivity may be adjusted by diafiltration against water, suitable diluent or buffer so that the process may run as a combination of ultrafiltration (UF) / diafiltration (DF) / ultrafiltration (UF) steps.
  • the starting material comprises the macromolecule and an organic polymer such as Pluronic F-68.
  • UF ultrafiltration
  • DF diafiltration
  • WO 2009/073569 A2 discloses an aqueous formulation comprising water and a protein, and methods of making the same.
  • the aqueous formulation of the invention may be a high protein formulation and/or may have low levels of conductivity resulting from the low levels of ionic excipients. It is also provided a method of preparing an aqueous formulation comprising a protein and water, the method comprising:
  • a conventional UF/DF process known from prior art as already described typically includes three steps.
  • This known 3-step UF/DF process is illustrated in Figure 2. It is shown a schematic representation of an UF/DF process for conditioning and concentrating a protein solution using two ultrafiltrations UF1 and UF2 and between these both ultrafiltration steps one diafiltration step DF1.
  • the three steps of Figure 2 are:
  • UF1 ultrafiltration concentrating the protein solution to, for example, a third to a half of the final target value
  • DF diafiltration, usually performed in several cycles thereof, against pure water to remove initial excipients
  • UF2 ultrafiltration to concentrate the protein solution to the desired final level.
  • the process developers and formulators have generally assumed that a) the excipient profile of the resulting solution will be well-defined, b) the final excipient profile will match that of the medium-exchange solution or diafiltration medium, and c) ultrafiltration will remove residual excipients while diafiltration will avoid residual carryover altogether.
  • the diavolume is the ratio between the total collected filtrate or permeate volume and the constant feed volume (Kurnik R.T., Yu A.W., Blank G.S., Burton A.R., Smith D., Athalye A.M., Van Reis R. (1995), Buffer exchange using size exclusion chromatography, countercurrent dialysis, and tangential flow filtration: Models, development, and industrial application, Biotechnology and Bioengineering, 45 (2), 149-157).
  • the sieving coefficient describes the ratio of solute concentrations in the filtrate and retentate. In the ideal case of free solute flow, the permeation of small molecules shows a linear solute decrease as a function of diavolumes on a logarithmic scale with a sieving coefficient reaching 1.
  • Equation 1 is typically used to determine the number of diavolumes or cycles required to reduce small molecules and impurities to a specified level (Harinarayan C, Skidmore K., Kao Y., Zydney A.L., Van Reis R. (2009), Small molecule clearance in ultrafiltration/diafiltration in relation to protein interactions: Study of citrate binding to a fab, Biotechnology and Bioengineering, 102 (6), 1718-1722). However, this equation does not consider non-equilibrium states.
  • c Ci [1 -exp (-N)] (2) where c, is the bulk concentration of component i added during diafiltration. S is assumed to be 1.
  • the estimated dissociation constants were in the millimolar range, and the strength of binding correlated with anion size and the number of ionized groups per molecule.
  • These investigations identified the binding sites as specific positively charged lysine amino acids on a single IL-1 ra cluster (Raibekas et al. loc. cit).
  • Other proteins - including calmodulin, lactic dehydrogenase, citrate synthase, fumarase, and malate dehydrogenase - are known to bind citrate (Neufeld T., Eisenstein M., Muszkat K.A., & Fleminger G. (1998), A citrate-binding site in calmodulin, Journal of Molecular Recognition 1 1 , 20-24).
  • Esue et al. Esue O., Kanai S., Liu J., Patapoff T.W., Shire S.J. (2013), Carboxylate-dependent gelation of a monoclonal antibody, Pharm. Res. 26 (2009) 2478-2485
  • Shao and Zydney (Shao J, Zydney A.L. (2004a), Optimization of ultrafiltration/diafiltration processes for partially bound impurities, Biotechnology and Bioengineering, 87 (3), 286-292; Shao J., Zydney A.L., (2004b), Retention of small charged impurities during ultrafiltration, Biotechnology and Bioengineering, 87 (1 ), 7-13) showed how binding interactions between impurities and large molecules (like proteins) significantly reduce impurity clearance rates. As a consequence, large increases in the number of diavolumes are required to obtain a given level of impurity removal. They proposed an analytical expression for calculating optimal diafiltration steps that accommodate protein-excipient interactions.
  • Harinarayan et al. found a specific electrostatic interaction between the tested antibody fragment (Fab) and trivalent citrate (while no interaction was observed between the Fab and a monovalent acetate) (Harinarayan et al. loc.cit.).
  • the Donnan effect thus plays an especially prominent role in membrane equilibrium and membrane potential of non-dialysable electrolytes (Brezesinski G., Mogel H.S., Grenzflachen und Kolloide, (1993), Physikalisch-chemische Kunststoffn, Spekrum Akademischer Verlag Heidelberg, Berlin, Oxford).
  • Various researchers have attempted to develop theoretical models incorporating the Donnan effect to better predict excipient and pH changes in protein solutions (e.g. monoclonal antibodies) (e.g.
  • the degree of carryover depends, for example, on both the pH of the initial biomolecule solution, biomolecule-excipient interaction and the number of diafiltration volumes. Diafiltration with pure water, even with a large number of cycles (approximately 20-25), could not eliminate carryover of anionic excipients like succinate and acetate. In practice, the number of diavolume exchanges necessary to clear the excipients substantially exceeded the number calculated from the above-mentioned Equation 1 (a mathematical model for calculating the clearance of small molecules of a diafiltration). Furthermore, one has to keep in mind, that increasing the diafiltration cycles may have a negative impact on protein integrity and increases process time.
  • a further object of the present invention is to provide a purer liquid biomolecule formulation product with the process, which may be purified easier and faster compared with the prior art processes, whereby the degree of carryover of excipients, particularly of anionic excipient(s), is reduced or even minimized.
  • a further object of the present invention is to provide a process, in which the number of diavolume exchanges necessary to clear the excipient(s) is in a reasonable range in order not to impair biomolecule stability and trigger aggregate formation.
  • a still further object of the present invention is to provide a process which is also feasible in large scale, provides the desired quality standard and operation efficiency with reasonable costs.
  • step (c) a second diafiltration DF2; and (d) a second ultrafiltration UF2; wherein an aqueous solution of one or more salts, as liquid medium B, is used for step (b) and water or an aqueous solution of one or more salts, as liquid medium C, is used for step (c), wherein the one or more salts used for step (b) are the same or different from the one or more salts used for step (c) and wherein the liquid medium B has an ionic strength which is higher than the ionic strength of the liquid medium C.
  • highly concentrated liquid formulation containing biomolecules in the frame of the present invention shall be understood in the sense that the biomolecule(s) is (are) present in the liquid formulation in a concentration of 70 mg/ml or more, or 80 mg/ml or more, or 85 mg/ml or more.
  • the process of the present invention was found to consistently reduce levels of undesired excipient(s) present in the starting liquid biomolecule formulation, preferably below the limit of detection, and resulted in stable liquid formulations of the biomolecules contained.
  • the biomolecules are selected to be proteins
  • the process yielded solutions with only enough exchange-medium counterions to balance proteins' inherent charge and permit the protein to self-buffer. Biomolecule integrity and biomolecule quality was generally found to be acceptable or completely unchanged.
  • any kind of unwanted excipient(s) present in the starting liquid biomolecule formulation can be removed with the process of the present invention performed under solution conditions such as positively charged excipient(s), negatively charged excipient(s) and neutral excipient(s).
  • a further advantage of the process according to the invention resides in the fact that it is very mild with regard to physical stress exerted onto the proteins. This can be concluded from a high degree of monomer content throughout the steps of the process.
  • liquid formulation for example, the expressions "liquid formulation”, “solution” “soluble” and “dissolved” or “solved” according to the present invention should be understood in their broadest meaning and include all kind of mixtures of a solid or liquid in a liquid medium such as true solutions, dispersions, suspensions and the like, unless otherwise stated.
  • the expression "highly concentrated” should be understood in the sense that the liquid biomolecule formulation is provided in a concentration which is higher than the starting concentration, preferably significantly higher than before. The exact increase of the concentration provided depends from each single case, the biomolecule and medium chosen as well as conditions and parameters of the ultrafiltration and diafiltration equipments used.
  • ultrafiltration refers to any technique in which a liquid formulation is subjected to a semi-permeable membrane that retains biomolecules, for example proteins, while allowing solvent and solute molecules smaller than the biomolecule to pass through.
  • ultrafiltration is used to increase the concentration of biomolecules, for example proteins, in a liquid formulation.
  • diafiltration or “DF” and similar terms refer to, for example, using a semi-permeable filtration membrane to remove, replace, or lower the concentration of salts or solvents from liquid formulations containing proteins, peptides, nucleic acids, or other biomolecules.
  • DF diafiltration membrane
  • diafiltration step refers to a total volume exchange (as far as possible) during the process of diafiltration.
  • DF/UF diafiltration/ultrafiltration
  • ultrafiltration and diafiltration are used sequentially.
  • excipient or “excipient(s)” or “excipients” refer in the present invention to one or more substances or compounds, such as auxiliary agents, ions, fragments or any kind of species that are present in the liquid biomolecule formulation besides the biomolecule itself and the solvent(s) used.
  • the excipient(s) present in the starting biomolecule formulation are excipient(s) which shall be reduced or removed as far as possible according to the process of the present invention. These excipient(s) may be charged or neutral in aqueous solution.
  • first excipient(s) or “starting excipient(s)”.
  • first or starting excipient(s) present in the starting liquid biomolecule formulation will be exchanged with other (second) excipient(s) which are more easily removable from the liquid biomolecule formulation, provide better compatibility or are more acceptable due to other reasons in order to obtain a well-defined liquid biomolecule formulation.
  • the second excipient(s) are used in step (b).
  • third excipient(s) to be used in step (c) may be optionally used.
  • fourth excipient(s) may be optionally used, i.e. excipient(s) which may be added after completion of the process according to the present invention. Even if the term "excipient" is used in singular it always comprises one or more excipients as the context may allow.
  • ionic excipient(s) refers to an ion(s) that has a net charge in an aqueous solution.
  • examples of ionic excipient(s) include, but are not limited to, anions derived from inorganic and/or organic salts, e.g. inorganic and/or organic buffering salts, or anions or cations, e.g. derived from detergents.
  • the ionic excipients may or may not interact with the biomolecule present.
  • salt refers to an ionic compound resulting from the neutralization of an acid and a base.
  • the salts are composed of positively charged ions, namely cations, and negatively charged ions, namely anions. These ions can be inorganic or organic.
  • An "organic salt” is therefore a compound wherein the cation and/or the anion is an organic compound.
  • acids and bases used are pharmaceutically acceptable also the salts thereof are pharmaceutically acceptable.
  • water is intended to mean any type of water which may be used.
  • Purified water may be preferred but according to some embodiments also tap water may be used.
  • the type of water selected depends from the intended use of the obtained liquid biomolecule formulation.
  • Purified water used according to the present invention is water that has been undergone a purification process such as distillation, reverse osmosis, carbon filtering, capacitive or electro-deionization, micro- or ultrafiltration, ultraviolet oxidation or the like to remove impurities to be suitable for use. Combinations of these processes may also be used in order to achieve water of such high purity, e.g. ultrapure water, that its trace contaminants are measured in parts per billion (ppb) or parts per trillion (ppt).
  • ppb parts per billion
  • ppt parts per trillion
  • water used in the process of the invention is ultrapure water, for example ultrapure water of type 1 , according to ASTM D1 193 or ISO 3696.
  • the water used may be sterile water suitable for administration to a subject such as water for injection (WFI).
  • WFI water for injection
  • distilled water, bidistilled or deionized water may be used.
  • exchange used throughout the present invention has to be understood in its broadest meaning which does usually not represent a complete exchange of one liquid medium containing excipient(s) against another liquid medium containing other excipient(s). It is rather a washing out or diluting of solvent and/or excipient(s).
  • the excipient succinate in water is washed out to be replaced with the excipient acetate in water (e.g. in step (b)).
  • a variety of other examples of exchanges exists.
  • a “stable” formulation according to the present invention is one in which the biomolecule, preferably protein, contained therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage.
  • process conditions and parameters for each individual step may vary depending on the particular biomolecules, excipient(s), mediums and filter systems employed. Unless otherwise specified, the process conditions and parameters of each process step may be readily selected by one of ordinary skill in the art. Exemplary procedures are provided in the Examples section.
  • biomolecule + solvent(s) biomolecule + solvent(s)
  • excipient(s) which is necessary, e.g. in view of the biomolecule used or for other reasons; e.g. ions such as counterions of the biomolecule may or must be present.
  • ions such as counterions of the biomolecule may or must be present.
  • it is provided a double u Itra -/d i af i I trati o n in the order of sequence step (a) - step (b) - step (c) - step (d) or ultrafiltration UF1/diafiltration DF1/ diafiltration DF2/ ultrafiltration UF2.
  • FIG. 3 a schematic representation of one exemplary embodiment of an UF/DF process according to the present invention for conditioning and concentrating a liquid biomolecule formulation using two ultrafiltration and two diafiltration steps is shown.
  • the legend is as follows:
  • CPI initial biomolecule concentration
  • the liquid biomolecule formulation is provided either as a commercially available product from a manufacturer on the market or it is prepared based on standard procedures known in prior art.
  • the biomolecule In the initial state the biomolecule is contained in a solvent or a mixture of solvents and is present in a concentration CPI.
  • the term "biomolecule" should per se encompass one or more biomolecules even if used in singular.
  • the starting liquid formulation contains a number of excipients (hereinafter also referred to as starting or first excipients), which may be generally indicated as excipients XYZ.
  • the solvent(s) and the excipient(s) contained in the starting liquid biomolecule formulation are hereinafter indicated as liquid medium A.
  • the (starting or first) excipients XYZ present are, for example, auxiliary agents used to stabilize, solubilise and/or formulate a biomolecule in the starting liquid formulation.
  • these excipients shall be reduced to a low level or even removed as far as possible in the final liquid formulation obtained because these excipients could negatively affect the performance, properties and behaviour of the liquid biomolecule formulation, for example, in further processing.
  • the excipient(s) XYZ present result in a formulation which may not be exactly defined which is not desirable at all.
  • the starting excipients may be in any form present in the liquid formulation such as a solid, complex, ion or the like dissolved or dispersed in the solvent(s) present.
  • excipient(s) are herein understood to mean only those excipient(s) which are present in any form in solution as defined above, because not dissolved and precipitated excipients may be separated easily from the liquid formulation.
  • the excipients XYZ are to be removed by the inventive process while the biomolecule(s) is(are) maintained in solution during the whole process in order to avoid potential biomolecule stress.
  • starting or first excipient(s) are not limited according to the present invention, they may be any kind of excipient(s) known in the art which are charged or neutral in aqueous solution.
  • the excipient(s) may be present in the starting liquid biomolecule formulation due to several reasons associated, for example, with the manufacturing, storage, pre-processing of the biomolecule, or properties of the biomolecule itself or the solvent(s) used or other reasons.
  • the excipient(s) to be reduced or removed as far as possible are the starting excipients which are charged or neutral in aqueous solution, for example, additives used in the preparation or processing of biomolecule; unwanted substances or compounds such as impurities contained in the starting liquid biomolecule formulation; undesired side-products formed during the manufacturing process of the biomolecule; decomposition or degradation products of starting, intermediate or biomolecule end products formed during the production of the biomolecule.
  • the excipient(s) might be cell components or debris, degradation products of bacteria such as endotoxines, DNA, RNA, undesired lipids, HCP (Host cell proteins), lipopolysaccharides (LPS) or parts thereof; sugars; detergents such as positively charged, negatively charged and also non-ionic species; any kind of negatively or positively charged ions preferably resulting from organic and/or inorganic salts, such as organic and/or inorganic buffer salts, and detergents.
  • endotoxines such as endotoxines, DNA, RNA, undesired lipids, HCP (Host cell proteins), lipopolysaccharides (LPS) or parts thereof
  • sugars such as positively charged, negatively charged and also non-ionic species
  • detergents such as positively charged, negatively charged and also non-ionic species
  • any kind of negatively or positively charged ions preferably resulting from organic and/or inorganic salts, such as organic and/or inorganic buffer salts, and detergents.
  • Charged excipients may be, for example, charged ions resulting from organic and/or inorganic salts dissolved in the aqueous solvent such as anions or cations, preferably anions.
  • the charged excipients may be derived from organic and/or anorganic buffer salts.
  • ions resulting from a buffer system used to stabilize the biomolecule starting or first ions
  • second ions may be replaced with other ions (second ions).
  • the starting excipient(s) to be removed may be understood as impurities of the liquid formulation. Impurities are usually present in small amounts but the impurities according to the present invention might be present in high quantities, for example, an anion of the buffer system. Therefore, these impurities are herein more correctly referred to as excipient(s) (starting or first excipient(s)).
  • the biomolecule and the starting excipient(s) have opposite charges.
  • the biomolecule may be positively charged such as in case of a protein and the excipients to be removed by the process are negatively charged excipients, such as anions.
  • Excipients which may be reduced or removed in this case may be, for example, buffering excipients such as citrate, succinate, acetate, and phosphate.
  • the focus of the present invention may be particularly directed to separate organic ions such as mono- or multivalent negative ions from liquid biomolecule formulations such as liquid protein formulations but not multivalent metal ions.
  • biomolecule(s) used according to the present invention is(are) not limited at all, any biomolecule(s) known by those skilled in the art may be used.
  • a biomolecule is any organic substance that is present in living organisms, including large macromolecules such as proteins, carbohydrates, lipids, and nucleic acids, as well as small molecules such as primary metabolites, secondary metabolites, and natural products. Most biomolecules in essential consist of the elements carbon, hydrogen, oxygen, nitrogen, and optionally phosphorous and sulfur. Also other elements may be present but only in small amounts. Biomolecules may be selected from small molecules, monomers, macromolecules and others. Exemplary small molecules are lipds such as phospholipids, glycolipids, sterols; vitamins; hormones; neurotransmitter.
  • Monomers which may be mentioned, but without restriction, are amino acids, nucleotides, monosaccharides etc.
  • Macromolecules or so-called biopolymers which may be used according to the present invention are, for example, proteins or peptides such as oligopeptides; nucleic acids such as DNA, RNA; oligosaccharides, polysaccharides such as glycogen, starch, chitin, cellulose, fructane, dextrane.
  • Particularly preferred biomolecules are biopolymers, particularly selected from proteins or peptides e.g. oligopeptides, nucleic acids, oligosaccharides, and polysaccharides. Most preferred are proteins or peptides.
  • polypeptide or "protein” are used interchangeably. These terms refer to polymers of amino acids of any length. These terms also include proteins that are post-translationally modified through reactions that include, but are not limited to glycosylation, glycation, acetylation, phosphorylation, oxidation, amidation or protein processing. Modifications and changes, for example fusions to other proteins, amino acid sequence substitutions, deletions or insertions, can be made in the structure of a polypeptide while the molecule maintains its biological functional activity. For example certain amino acid sequence substitutions can be made in a polypeptide or its underlying nucleic acid coding sequence and a protein can be obtained with similar or modified properties. Amino acid modifications can be prepared for example by performing site-specific mutagenesis or polymerase chain reaction mediated mutagenesis on its underlying nucleic acid sequence.
  • polypeptide and protein thus also include, for example, fusion proteins consisting of an immunoglobulin component (e.g. the Fc component) and a growth factor (e.g. an interleukin), antibodies or any antibody derived molecule formats or antibody fragments.
  • an immunoglobulin component e.g. the Fc component
  • a growth factor e.g. an interleukin
  • the term “protein” or “polypeptide” includes proteins, polypeptides, fragments thereof, peptides, fusion proteins all of which can be expressed in the selected host cell.
  • the protein is a recombinant protein, i.e., a protein encoded by a recombinant DNA resulting from molecular cloning.
  • proteins can be antibodies, enzymes, cytokines, lymphokines, adhesion molecules, receptors and derivatives or fragments thereof, and any other polypeptides that can serve as agonists or antagonists and/or have therapeutic or diagnostic use or can be used as research reagent.
  • the protein is a secreted protein or protein fragment, more preferably an antibody or antibody fragment or an Fc-fusion protein.
  • RNA may also be an antisense RNA, tRNA, rRNAs, other RNAs being part of riboproteins or other regulatory RNAs.
  • antibody antibodies
  • immunoglobulins There are various classes of immunoglobulins: IgA, IgD, IgE, IgG, IgM, IgY, IgW.
  • the antibody is an IgG antibody, more preferably an lgG1 antibody.
  • Antibody includes a polyclonal, monoclonal, monospecific, bi-specific, multi-specific, a single chain antibody, an antigen-binding fragment of an antibody (e.g., an Fab or F(ab')2 fragment), a disulfide-linked Fv, etc.
  • Antibodies can be of any species and include chimeric and humanized antibodies.
  • “Chimeric” antibodies are molecules in which antibody domains or regions are derived from different species. For example the variable region of heavy and light chain can be derived from rat or mouse antibody and the constant regions from a human antibody. In "humanized” antibodies only minimal sequences are derived from a non-human species. Often only the CDR amino acid residues of a human antibody are replaced with the CDR amino acid residues of a nonhuman species such as mouse, rat, rabbit or llama. Sometimes a few key framework amino acid residues with impact on antigen binding specificity and affinity are also replaced by non-human amino acid residues. Antibodies may be produced through chemical synthesis, via recombinant or transgenic means, via cell (e.g., hybridoma) culture, or by other means.
  • Immunoglobulins are tetrameric polypeptides composed of two pairs of a heterodimer each formed by a heavy and light chain. Stabilization of both the heterodimers as well as the tetrameric polypeptide structure occurs via interchain disulfide bridges.
  • Each chain is composed of structural domains called “immunoglobulin domains” or “immunoglobulin regions” whereby the terms “domain” or “region” are used interchangeably.
  • Each domain contains about 70 - 1 10 amino acids and forms a compact three-dimensional structure.
  • Both heavy and light chain contain at their N-terminal end a "variable domain” or “variable region” with less conserved sequences which is responsible for antigen recognition and binding.
  • the variable region of the light chain is also referred to as "VL” and the variable region of the heavy chain as "VH”.
  • Fc fragment crystallizable
  • These may be formed by protease digestion, e.g. with papain or pepsin from conventional antibodies but may also be produced by genetic engineering.
  • the N-terminal part of the Fc fragment might vary depending on how many amino acids of the hinge region are still present.
  • Fc-fusion protein describes polypeptides which contain as a fusion partner a natural or modified (e.g. substitutions, deletions, insertions) Fc region of an immunoglobulin.
  • Fc fusion proteins can be either naturally occurring proteins (e.g. antibodies) or engineered recombinant proteins (e.g. TNF receptor-Fc fusion protein or a VH region fused to an Fc region).
  • the Fc-fusion proteins can exist either as monomers or as multimers whereby polypeptides can have identical or different sequences, might contain linker sequences between the two fusion partners and/or part of the hinge region or modified hinge regions or the polypeptide is fused directly to the CH2 domain.
  • an antibody protein of this kind is known as a "single-chain-Fv" or "scFv”.
  • scFv-antibody proteins of this kind are known from the prior art.
  • more than one VH and/or VL region can be linked together.
  • scFv as a multimeric derivative. This is intended to lead, in particular, to recombinant antibodies with improved pharmacokinetic and biodistribution properties as well as with increased binding avidity.
  • scFv were prepared as fusion proteins with multimerisation domains.
  • the multimerisation domains may be, e.g. the CH3 region of an IgG or coiled coil structure (helix structures) such as Leucine-zipper domains.
  • the interaction between the VHA L regions of the scFv is used for the multimerisation (e.g. dia-, tri- and pentabodies).
  • diabody By diabody the skilled person means a bivalent homodimeric scFv derivative.
  • Diabodies may additionally be stabilized by the incorporation of disulphide bridges. Examples of diabody-antibody proteins are known from the prior art.
  • minibody means a bivalent, homodimeric scFv derivative. It consists of a fusion protein which contains the CH3 region of an immunoglobulin, preferably IgG, most preferably lgG1 as the dimerisation region which is connected to the scFv via a Hinge region (e.g. also from lgG1 ) and a linker region.
  • an immunoglobulin preferably IgG
  • lgG1 as the dimerisation region which is connected to the scFv via a Hinge region (e.g. also from lgG1 ) and a linker region.
  • Hinge region e.g. also from lgG1
  • linker region e.g. also from lgG1
  • triabody By triabody the skilled person means a: trivalent homotrimeric scFv derivative. ScFv derivatives wherein VH-VL is fused directly without a linker sequence lead to the formation of trimers.
  • miniantibodies which have a bi-, tri- or tetravalent structure and are derived from scFv.
  • the multimerisation is carried out by di-, tri- or tetrameric coiled coil structures.
  • a nanobody also known as single-domain antibody is an antibody fragment consisting of a single monomeric variable antibody domain. Like a whole antibody, it is able to bind selectively to a specific antigen.
  • Nanobodies have a molecular weight of about 12-15 kDa, and are therefore much smaller than common antibodies having molecular weigths in the range of 150 to 160 kDa, which are composed of two heavy protein chains and two light chains. They are also smaller than Fab fragments (about 50 kDa) and single-chain variable fragments (about 25 kDa).
  • the term “humanantibody derived molecule(s)” is used interchangeably with “antibody derived fragments” or “antibody fragments” and refers to polpypeptides which contain only part(s) of one or more antibody domain(s) or region(s) and/or complete domain(s) or region(s).
  • the antibody fragments can be either a) forming a molecule on their own, b) linked with each other in different combinations, c) fused to non-antibody sequences, d) fused or linked to non-polypeptide (e.g. radionucleotides) or d) any combination of the above.
  • These polypeptides can exist either as monomers or as multimers whereby polypeptides can have identical or different sequences.
  • proteins to be used per se as biomolecule shall not be limited to the use in the pharmaceutical and biotechnology sectors but any kind of protein in any type of application field can be used. Proteins are also known to be used in a variety of non-pharmaceutical applications, for example in the food stuff industry, animal feed industry, textile industry, chemical-technical industry, detergent industry and other sectors.
  • Preferred proteins are therapeutic proteins, non-therapeutic proteins, antibodies, antigen-binding fragments or nanobodies, particularly monoclonal antibodies and related compounds or formates.
  • the process of the present invention may be employed in a particular advantageous manner if the biomolecules are positively charged and the excipient(s) (starting or first excipients) to be removed by the process are negatively charged excipient(s).
  • step (a) a first ultrafiltration (UF1 ) is performed by which the biomolecule containing liquid formulation is concentrated and a concentration up to CP2 of the biomolecule is reached. That is, the ultrafiltration UF1 of step (a) is used to concentrate the liquid biomolecule formulation, preferably up to about 10%-70%, or, more preferably about 15%-60%, or, most preferably about 25%-50% compared with the initial concentration of the liquid biomolecule formulation.
  • This kind of concentration has the advantage to reduce the overall process volume for the next DF step and this also leads to reduced process times.
  • the ultrafiltration as well as the subsequent diafiltrations selectively utilizes permeable (porous) membrane filters to separate the components of the liquid formulation based on their molecular size.
  • a membrane retains biomolecules that are larger than the pores of the membrane while smaller molecules such as salts, ions, solvents such as water, which are permeable, freely pass through the membrane.
  • One parameter for selecting a membrane is its retention characteristics. As a general rule, the molecular weight cut-off (MWCO) of the membrane should be 1/3 to 1/6 the molecular weight of the biomolecule to be retained. It is a matter of course that the excipient(s) to be removed have a lower or even significantly lower molecular weight than the biomolecule so that the biomolecule is retained and not the excipient(s).
  • the liquid biomolecule formulation used in step (a) contains a liquid medium A composed of solvent(s) and starting or first excipient(s), whereby the medium A is exchanged in step (b) with medium B, and medium B is exchanged in step (c) with medium C, whereby the liquid biomolecule formulation primarily containing liquid medium C (and probably small amounts of liquid mediums B and C) results, which has a reduced content of the starting excipient(s), being preferably lower than the level of detection.
  • medium A contains or consists of solvent(s) and undesired starting or first excipient(s), which are planned to be exchanged/replaced, whereby medium A is exchanged by means of liquid medium B against liquid medium C.
  • Liquid medium B contains or consists of solvent(s) and second excipient(s)
  • medium C contains or consists of solvent(s) and third excipient(s), preferably solvent(s) only.
  • the second excipient(s) and optional third excipient(s) are the same or different from each other; both are different or at least partially different from the first excipient(s) which shall be removed. "At least partially different" has to be understood in the sense that e.g. one or more of the first excipients may be the same with one or more of the second excipients.
  • sodium chloride may be present as first excipient and also as second excipient. This does not interfere with the process to be performed. However, there is always a difference between first and second excipients in total in order to result in a real exchange of first excipient(s) by second excipient(s).
  • the type of the excipient is not of particular importance because any type of excipient whether charged or neutral may be removed.
  • the invention provides the possibility to remove any type of excipient(s) although strong biomolecule-excipient(s) interactions being present.
  • the first excipient is an acetate buffer salt which is exchanged with a second excipient such as a succinate buffer salt, and the second excipient is exchanged with a third excipient e.g. chloride.
  • a second excipient such as a succinate buffer salt
  • a third excipient e.g. chloride
  • the liquid medium C consists or essentially consists of water.
  • the first excipient may be succinate, which is replaced by acetate as second excipient and so on.
  • the "second excipients” and “third excipients” are herein used interchangeably with “salts” whereas the “starting or first excipients” and also “fourth excipients” (which may be added to the obtained final liquid biomolecule formulation) are not only salts but may be also other substances or compounds as herein described, for example any kind of suitable auxiliary agents such as detergents, surfactants, sugars etc.
  • the starting or first excipient(s) present in the starting liquid biomolecule formulation are excipient(s) which are not desired to be present and shall be removed by the process according to the present invention.
  • the liquid medium B used in step (b) has an ionic strength which is higher than the ionic strength of the liquid medium C used in step (c).
  • Mediums B and C may be selected from any liquid medium which may be used in connection with a biomolecule, which may be able to maintain the biomolecule in a liquid formulation and do not have any negative influence on the characteristics of the biomolecule used. It is a matter of course that the liquid mediums B and C (and A) used should not in any way transform or alter the biomolecule contained in it.
  • the only interaction which may be accepted is an ionic interaction of the ions contained in the liquid medium with the biomolecule in order to stabilize it. This is in case the biomolecule and the liquid medium have opposite charges.
  • the biomolecule is a positively charged biomolecule such as a protein and the anions of the liquid medium interact with the protein to stabilize it in solution.
  • Mediums B and C and preferably also medium A represent an aqueous solution. Therefore, the solution always contains water. Further, the solution contains a solvent or a mixture of solvents, particularly the solution contains water and another solvent or water and a mixture of solvents.
  • the solvent(s) may be selected from any known solvent which is miscible with water and does not in any manner adversely affect the properties of the solved biomolecule such as an antibody or nanobody. In case the biomolecule provided is intended for pharmaceutical use it is a matter of course that the solvent(s) selected shall be likewise suitable for pharmaceutical use, too.
  • the solvent or solvents is/are preferably selected from the group consisting of mixtures of water and organic solvent(s) miscible with water.
  • organic solvent(s) may be exemplarily mentioned but not limited to alcohols such as ethanol, methanol, glycols, sugar alcohols, e.g. glycerine, acetone, acetonitrile, methyl ethyl ketone, ethers (those which are miscible with water) such as dioxan, diglyme, dimethylformamide, N-methyl- pyrrolidone, tetrahydrofuran, and the like and mixtures thereof.
  • the type of solvent(s) used strongly depends from the biomolecule(s) present and the intended use of the end product.
  • the solution may contain predominantly organic solvent(s); however, it may be preferred if the solvent is predominantly composed of water, so that water represents the main part of the solvents present. According to a further preferred embodiment the solvent used may consist or essentially consist of water alone.
  • the solvent(s) used in liquid mediums A, B and C may be the same or different. According to a preferred embodiment the solvent(s) used is(are) the same in all liquid mediums A, B, and C. According to a particularly preferred embodiment the solvent is water.
  • liquid medium B and/or C (and also liquid medium A) aqueous salt solutions are used
  • organic salt solutions and/or inorganic salt solutions in water may be exemplarily mentioned.
  • the organic and/or inorganic salts used are preferably water soluble and completely inert with regard to the biomolecule used.
  • liquid medium B comprises sodium chloride in a concentration from about 150 to about 900 mM, increasingly preferred from about 200 to about 700 mM, from about 400 to about 600 mM, and from about 450 to aboput 550 mM. This mode is especially preferred when liquid medium B does not comprise a specific low molecular weight buffering agent.
  • Such mode is advantageous in view of the pH values of the subsequent liquid media.
  • the pH value of such process step (b) and/or one or more of the following steps varies only in minor ranges, i.e. less than about 0.4, preferably less than about 0.3 pH, even more preferred less than about 0.2 pH values.
  • Examples 3, 6, 9, 10, 1 1 , 12, and 13 exemplify this mode with 200 and 500 mM NaCI and a resulting pH of the liquid medium D regularly just about 0 to 0.3 pH values below the pH value of liquid medium B, i.e. slightly more acidic.
  • This mode of the invention is especially useful for biomolecules which are sensitive against stronger pH variations.
  • the organic and inorganic salts used are not limited according to the present invention if they may be used to provide a liquid formulation of the biomolecule present.
  • the salts used in the liquid medium B and/or C may be organic and/or inorganic salts, preferably organic and/or inorganic buffer salts, e.g. usable as biological buffers.
  • a buffer is a combination of a weak acid or a weak base and its conjugate salt form that keeps the pH from shifting out of the optimal range when added acid or base gets into the system. It is a matter of course that the buffer in the liquid medium used should not in any way react or alter or negatively interact with the biomolecule contained in it, except for the allowed ionic interactions between biomolecule and counterions.
  • buffers used are freely soluble in water and poorly soluble in other solvents and they represent an inert system.
  • the aqueous solution in form of the liquid mediums B and or C represents or may contain a buffer which is on basis of water as solvent or represents or may contain a buffer which is on basis of another solvent or solvents but contains water.
  • the organic acids and bases to prepare the buffer or the buffers per se are not limited according to the frame of the present invention but any acid, base or buffer usable in connection with the biomolecule selected may be used.
  • the buffer shall be also selected from pharmaceutically acceptable buffers, e.g. biological buffers.
  • the buffer may be, for example, selected from one or more pharmaceutically acceptable or compatible buffers or buffering agents.
  • so-called biological buffers may be used, i.e. buffers which are known from prior art to be used for biological system or in the context thereof.
  • Exemplary biological buffers which may be used in or as liquid medium A, B, and C according to the present invention may be listed as follows but without limitation to the mentioned specific examples:
  • Possible buffers or buffer salts are on basis of N-(2-acetamido)- aminoethanesulfonic acid (ACES) and salts thereof, acetic acid and salts thereof, aconitic acid and salts thereof, adipic acid and salts thereof, ascorbic acid and salts thereof, A/-(2-Acetamido)-iminodiacetic acid (ADA) and salts thereof, ammonia and salts thereof, ammonium chloride, 2-amino-2-methyl-1 -propanol (AMP), 2-amino-2- methyl-1 ,3-propanediol, ammediol (AMPD), N-(1 ,1 -Dimethyl-2-hydroxyethyl)-3- amino-2-hydroxypropanesulfonic acid (AMPSO) and salts thereof, N,N-b ' ⁇ s- ⁇ 2- hydroxyethyl)-2-aminoethanesulfonic acid (BES) and salts thereof, benzoic acid and
  • buffers mentioned above are derived from organic salts
  • buffers on basis of inorganic salts may per se be used, such as phosphate buffers, for example, potassium hydrogen phosphate buffers and the like.
  • mixed buffers containing inorganic and organic salts may be used.
  • Further usable organic salts (inner salts) which are at the same time buffers, particularly biological buffers, are amino acids in aqueous solution.
  • Amino acids which may be used are, for example, alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine; particularly preferred amino acids are alanine, arginine, glutamine, glycine, histidine, lysine, and proline.
  • a buffer is a mixture of substances, i.e. a mixture of a weak base and the strong conjugated acid or a weak acid and the strong conjugated base, however, the buffer may be indicated in the present invention with reference to the acid or base or its respective conjugate salt form, only.
  • an "acetate” or “acetate buffer” shall be understood as a buffer which contains acetic acid and acetate salt(s). Those skilled in the art may readily understand the context in which it is referred to a buffer system and which components are contained in it.
  • Preferred buffers which may be used as or in medium B and/or C, are exemplarily selected, but without limitation, from the group consisting of phosphoric acid and salts thereof, citric acid and salts thereof, tris, succinic acid and salts thereof, malic acid and salts thereof, tartaric and salts thereof, acetic acid and salts thereof, lactic acid and salts thereof, aconitic acid and salts thereof, ascorbic acid and salts thereof, glutamic acid and salts thereof, ammoniumchloride, triethanolamine, alanine, arginine, glutamine, glycine, histidine, lysine, and proline.
  • a buffer is understood according to the present invention to be composed of a solvent(s) and inorganic and/or organic salt(s), the salt(s) being present in form of dissolved ions which are herein also referred to as excipient(s).
  • organic and/or inorganic buffer salt(s) instead of organic and/or inorganic buffer salt(s) also other salts may be used.
  • any inorganic salt(s) may be used.
  • the inorganic salt is not limited according to the present invention, any inorganic salt which is soluble in an aqueous solution and does not interfere with the biomolecule used may be employed.
  • the inorganic salt is for example selected from the group consisting of alkali salts or alkaline earth salts of sulfates, nitrates, phosphates, carbonates, halogenides, borates, silicates and the like.
  • the inorganic salt as well as the organic salt shall be selected from the group of pharmaceutically acceptable salts per se known.
  • pharmaceutically acceptable inorganic salts are selected from sodium salts such as sodium halides, preferably sodium chloride, sodium sulfate, sodium borate; calcium salts such as calcium halides, preferably calcium chloride, calcium sulfate, calcium borate; magnesium salts such as magnesium halides, preferably magnesium chloride, magnesium sulfate, magnesium borate, and combinations thereof as well as other pharmaceutically acceptable inorganic salts.
  • a particularly preferred inorganic salt is sodium chloride due to its advantages properties.
  • sodium chloride has only a minor influence on the pH value and is present in a number of known buffer systems.
  • a biomolecule will not be affected by sodium chloride and it is known to be harmless for animal and human.
  • a salt resulting in any type of charged ion in aqueous solution such as any mono- or multivalent ion may be used.
  • mono-, divalent or trivalent ions may be used.
  • monovalent ions may be preferably used.
  • liquid medium B has an ionic strength which is higher than the ionic strength of liquid medium C.
  • This requirement is an essential feature of the process provided in order to achieve the desired results.
  • the ionic strength of a solution is a measure of the concentration of ions in the solution. The ionic compounds dissolve in water, dissociate into ions and result in an ionic strength of the solution which is a function of the concentration of all ions present:
  • Equation c is the molar concentration of the ion i (M, mol/L), z, is the charge number of that ion, and the sum is taken over all ions n in the solution.
  • ionic strength is equal to the concentration, but for salts such as MgS0 4 the ionic strength is four times higher so that multivalent ions contribute strongly to the ionic strength.
  • a desired ionic strength of a salt solution for example a buffer
  • the setting of the ionic strength of a salt solution can be done depending on the concentration and ionic potency of the salts present.
  • a vast number of publications and patent documents exist in prior art so that a specific value or range of the ionic strength may be looked up in a handbook, a monograph or the like. Therefore, the skilled person is able to provide a salt solution which has the required ionic strength.
  • liquid medium B has a high ionic strength expressed in form of a concentration of at least about 20 mM or more, or, preferably, at least about 100 mM or more, or, most preferred, at least about 200 mM or more.
  • liquid medium C has a low ionic strength expressed in form of a concentration of about 150 mM or lower, or, preferably about 100 mM or lower, or, more preferably about 75 mM or lower, or, most preferred about 50 mM or lower .
  • the liquid medium B used in diafiltration DF1 has a higher ionic strength compared with the liquid medium C used in diafiltration DF2.
  • the ionic strength of liquid medium B indicated in form of a concentration may be in the range of from about 20 mM up to the limit of solubility of the salt, or, particularly preferred, from about 100 mM to 1000 mM, or, more preferred about 150 mM to 750 mM, or, most preferred from about 200 mM to 500 mM.
  • the limit of solubility shall be understood as the maximum solute concentration that can be dissolved at a given temperature.
  • the extent of the solubility of a substance in a specific solvent is e.g. measured as the saturation concentration, where adding more solute does not increase the concentration of the solution and begins to precipitate the excess amount of solute.
  • the limit of the solubility or quantitative solubility of a salt is the maximum concentration of the salt in the solvent(s) resulting in a system having just one phase.
  • the limit of solubility is dependent from temperature, pressure and the pH of the solution, the skilled person will be able to select and adjust the desired concentration based on the values known from prior art.
  • sodium chloride may be used in an ionic strength given as concentration from about 20 mM up to about 6 Mol/I.
  • the ionic strength of liquid medium C (expressed as concentration) may be in the range of from about 0 mM to 150 mM, or, particularly preferred, from about 0 mM to 100, or, more preferred, about 0 mM to 75 mM, or, most preferred from about 0 mM to 50 mM.
  • an aqueous solution of one or more salts as liquid medium B is used for step (b) and water or an aqueous solution of one or more salts as liquid medium C is used for step (c), and the liquid medium B has an ionic strength which is higher than the ionic strength of the liquid medium C.
  • the difference between the ionic strength of the liquid medium B and the ionic strength of the liquid medium C indicated as concentration is preferably at least about 100 mM, or, more preferred at least about 200 mM, or, most preferred at least about 500 mM.
  • the ionic strength of the liquid medium B is selected to be about 500 mM
  • the ionic strength of the liquid medium C is lower than about 500 mM, preferably selected to be about 400 mM or lower, or, more preferred, about 300 mM or lower, or, most preferred, about 0 mM.
  • water is a liquid medium C that has an ionic strength of about 0 mM and also an electric conductivity which is about 0 mS/cm.
  • the starting liquid medium A preferably has a lower ionic strength than liquid medium B. But this is not in any case necessary.
  • step (b) the process of the present invention may be adjusted accordingly in that the cycle number in step (b) and optionally also step (c) may be preferably increased, respectively.
  • step (c) the process of the present invention may be adjusted accordingly in that the cycle number in step (b) and optionally also step (c) may be preferably increased, respectively.
  • the skilled person is readily able to optimize the process steps accordingly.
  • the ionic strength of liquid medium A is higher than the ionic strength of liquid medium B but also the reverse is possible.
  • the liquid medium A may be provided with an ionic strength being in the suitable range, which may be controlled by the concentration step (a). It is also possible that the liquid medium A may be diluted with water in order to obtain a lower ionic strength but this is not preferred.
  • the liquid medium C used for step (c) (DF2) is water.
  • any water to be used during the whole process, per se or as aqueous solution in a medium should be very pure water in order to avoid a contamination of the liquid biomolecule formulation with ions contained in the water as already explained. Therefore, it is useful to employ ultrapure water, for example ultrapure water of type 1 , according to ASTM D1 193 or ISO 3696. However, for other non-pharmaceutical uses also tap water may be used.
  • the medium or solvent exchanging steps in the process according to the present invention take place via a liquid medium having high ionic strength (DF1 ) to a medium having low ionic strength (DF2) in order to remove the undesired excipients present.
  • DF1 liquid medium having high ionic strength
  • DF2 medium having low ionic strength
  • the transfer from higher ionic strength to lower ionic strength weakens the biomolecule- excipient(s) interactions so that these excipient(s) may be reduced or removed as far as possible.
  • the electrical conductivity which is associated with the ionic strength may be employed to determine the mediums used.
  • An empirical method relies on a simplified linear relationship between electrical conductivity and the ionic strength.
  • a liquid medium B having an ionic strength in the range from 200 mM to 500 mM approximately has an electrical conductivity in the range from 10 mS/cm to 50 mS/cm
  • a low electrical conductivity may be indicative that the liquid formulation has significantly reduced excipients including ionic excipients so that the conductivity may be used to determine the purification content obtained.
  • the parameters and conditions of the ultrafiltration and diafiltration steps which are per se known from prior art, can be readily selected and adapted accordingly by the skilled person.
  • the pH value of the liquid mediums A, B, C is not an issue in the process of the invention because the process functions at any pH value.
  • the pH value may be of interest if a charged ion is only present in dependence from the pH adjusted.
  • the acetate anion is only present in the chemical equilibrium acetate/acetic acid if the pH > 3.75. Therefore, it is a matter of course to perform the process in a suitable pH value or range where the salt or ion to be used to perform an exchange is present with the suitable charge.
  • both diafiltrations are consecutively performed in the same diafiltration system with the same separation filter but different mediums B and C.
  • Other embodiments are possible.
  • the first diafiltration (DF1 ) of step (b) may be repeated several times prior to performing the subsequent step (c). That is the exchange of liquid medium B may be performed for a number of times in the form of medium cycles as may be seen in Figure 1A.
  • step (b) is repeated 2 times, the cycle of figure 2A is passed 2 times and for any cycle the same liquid medium B is added as diafiltration medium with each cycle.
  • the first diafiltration DF1 which is preferably carried out at constant retentate volume, is therefore preferably performed against medium B in an amount to at least about two times the volume of medium B up to an amount of 10 times the volume of medium B.
  • the process step (b) is performed with at least a determined volume exchange, for example a 2-fold volume exchange with liquid medium B.
  • the second diafiltration (DF2) of step (c) may be repeated several times prior to performing the subsequent step (d). That is the exchange of liquid medium C may be performed for a number of times in the form of medium cycles as may be seen in Figure 1A.
  • step (c) is repeated 4 times, the cycle of Figure 2A is passed 4 times and for any cycle the same liquid medium C is added as diafiltration medium with each cycle.
  • the second diafiltration DF2 which is preferably carried out at constant retentate volume, is therefore preferably performed against medium C in an amount to at least about two times the volume of medium C up to an amount of 10 times the volume of medium C.
  • the process step (c) is performed with at least a determined volume exchange, for example a 2-fold volume exchange with liquid medium C.
  • the number of exchange volumes or exchange cycles highly depends on the diafiltration medium used, for example the ionic strength or concentration used. The skilled person is readily able to find out the optimum number of cycles of step (b) and step (c), respectively, by routine experimentation.
  • the second diafiltration DF2 is performed in accordance with the process of the invention using water alone as the diafiltration medium C.
  • step (c) DF2
  • a conductive salt(s) such as sodium chloride may be present to control the electrical conductivity. If only water is present the electrical conductivity is 0 mS/cm so that no measurable value is obtained. This is due to the fact that some UF/DF systems are run using conductivity probes. Therefore a small amount of e.g. 0.001 to 0.003 weight% of conductive salt is preferably added during or after step (c) (DF2) in order to better control the process.
  • the liquid medium C essentially consists of water due to the presence of a small amout of conductive salt(s).
  • liquid mediums B and C are the same and differ only with regard to the ionic strength used.
  • the conductive salt(s) represent the second and third excipient(s), only present in different concentrations.
  • a second ultrafiltration (UF2) is performed in order to obtain the liquid biomolecule formulation in concentrated form and a concentration up to CP3 of the biomolecule is reached.
  • the ultrafiltration UF2 of step (d) is used to concentrate the liquid biomolecule formulation to the desired value.
  • the ultrafiltrations according to step (a) and (d) can be accomplished with the same ultrafilter membrane.
  • the ultrafiltration steps may be performed with any suitable ultrafilter apparatus or ultrafilter membrane known.
  • the process of the present invention may be used to remove negatively charged excipient(s) from the starting liquid medium A containing the biomolecule(s) as positively charged compounds.
  • a part of the negatively charged excipient(s) may be used to stabilize the biomolecule in the liquid formulation.
  • a positively charged protein may be stabilized by the presence of anions. With the process of the present invention the anions will be reduced or removed as far as possible or necessary.
  • the anions (starting or first excipients) present in liquid medium A to stabilize the biomolecule will be replaced with anions (second excipients) present in liquid medium B so that the kind of anions in step (b) will represent the counterions of the biomolecule.
  • the liquid medium C (third excipients, if present) having a lower ionic strength than liquid medium B will usually not result in an exchange of the anions (second excipients) already present in step (b) as counterions of the biomolecule.
  • the anions (second excipients) present in liquid medium B will determine the counterions of the biomolecule so that the counterions may be selected accordingly.
  • the excipients of liquid medium B are the excipients which form a biomolecule- excipient complex obtained in step (d) of the inventive process.
  • the process of the present invention performed under solution conditions it is possible to use any excipient(s) for the first, second or third excipients such as positively charged excipient(s), negatively charged excipient(s) and neutral excipient(s). It is a matter of course that if the first or starting excipient(s) have a specific charge that also the second excipient(s) have the same corresponding charge in order to replace the first excipient(s) if this is required for any reason.
  • the first or starting excipient(s) have a specific charge that also the second excipient(s) have the same corresponding charge in order to replace the first excipient(s) if this is required for any reason.
  • the first excipient(s) are negatively charged, interact with the biomolecule and shall be replaced, then also the second excipients have a negatively charge etc.
  • the second and optional third excipients may be selected depending from the biomolecules, solvents and first excipient(s) present.
  • the second and third excipients are selected to be salts, preferably organic and/or inorganic salts, more preferably organic and/or inorganic buffer salts or ions derived thereof as herein described.
  • the excipient(s) present in liquid medium A are reduced as far as possible or required. According to a preferred embodiment the excipient(s) present in starting liquid medium A are reduced to be lower than the level of detection.
  • LOD Level of Detection
  • LOQ Level of Quantification
  • LOD 0.01 mM
  • the process steps (a) to (d) are performed at room temperature (20 - 25°C).
  • room temperature should be avoided throughout the whole process due to the temperature sensibility of a number of biomolecules to be used.
  • Temperatures in the range of about 2 to 35°C, preferably about 5 to 25°C, most preferably about 20 to 25°C are usually acceptable. For example, heating should not be performed at all during the whole process.
  • step (a) is step 1 or the first step to be performed in the process
  • step (b) is step 2 of the process
  • step (c) is step 3 of the process
  • step (d) is step 4 of the process. Another sequence of order is not intended and not desired.
  • step (b) and step (c) follow directly one after the other whereby no intermediate process step is performed in between; i.e. diafiltration DF2 follows directly after diafiltration DF1 without any intermediate step in between.
  • step (a) and step (b) follow directly one after the other whereby no intermediate process step is performed in between.
  • step (c) and step (d) directly follow one after the other whereby no intermediate process step is performed in between. Therefore, the steps (a) to (d) are preferably performed with no intermediate step in between, i.e. step (b) follows directly after step (a), step (c) follows directly after step (c) and step (d) follows directly after step (c).
  • the invention is also directed to a highly concentrated liquid formulation containing biomolecules prepared by a process as described above.
  • a well-defined liquid biomolecule formulation which may be used in the pharmaceutical or non- pharmaceutical field or as a good starting point for the development of pharmaceutical or non-pharmaceutical compositions.
  • the process of the present invention may be used to create a liquid biomolecule formulation to which defined excipients (fourth excipient(s)) may be added back in precise amounts allowing to provide a biomolecule formulation with precise concentrations of contents.
  • fourth excipient(s) are those already described above or known and described in prior art to be useful in liquid biomolecular formulations.
  • an improved and modified UF/DF process for preparing clearly defined biomolecule formulations may be provided, preferably clearly defined protein formulations may be obtained. It has been found that also with regard to problematic high biomolecule concentrations (such as 70 mg/ml_ or more) the process of the invention is smoothly functioning.
  • the process provides a final formulation consisting or essentially consisting solely of a highly concentrated biomolecule solution and a reduced amount of impurities, i.e. a residue of remaining excipients such as counterions of the biomolecule, if still present.
  • the process according to the present invention allows the preparation of well- defined highly concentrated formulations containing biomolecules, particularly proteins, intended for pharmaceutical or non-pharmaceutical use.
  • the obtained biomolecule formulation is provided as pure product formulation, which may be purified easier and faster compared with the prior art processes.
  • the number of diavolume exchanges necessary to clear the starting excipients is in a reasonable range so that biomolecule stability is not affected and aggregate formation not supported.
  • a further advantage of the process according to the invention is that the process is very mild with regard to physical stress exerted onto the proteins. This can be concluded from a high degree of monomer content which is present throughout the steps of the process.
  • the final product of the process according to the present invention is a clearly defined biomolecule formulation the further processing thereof is more simple and straightforward.
  • tailored formulations may be provided in an easier manner because the user can add the excipients of choice (fourth excipient(s)) by spiking additives to arrive at the desired defined formulation.
  • the process of the present invention is also feasible in large scale, provides the desired quality standard and operation efficiency with reasonable costs.
  • an effective UF/DF process which allows the removal of excipients under solution conditions up to very low levels, preferably lower than the level of detection.
  • the inventive UF/DF process may be also used in case of charged excipients and charged biomolecules.
  • negatively charged excipients such as citrate, succinate, and phosphate
  • Preferred charged biomolecules are e.g. proteins, such as antibodies which may be positively charged. Therefore, the diafiltrations allow users to condition the liquid biomolecule formulation and to replace one solution with another, by removing excipients in form of impurities, for example, remaining from preparation conditions such as fermentation or excipients required for measurement methods such as chromatography.
  • the process according to the present invention results in a formulation, achieved with repeated ultrafiltration concentrations and diafiltration washings which have cleared impurities or reduced them to levels that will not affect the safety, efficacy, or storage of the final product.
  • the double-diafiltration UF/DF process - incorporating two diafiltration steps, one at high ionic strength and one at low ionic strength (such as pure water) - shows that it is possible to obtain concentrated biomolecule formulations, preferably protein formulations whereby for example, anionic excipients such as phosphate, citrate, succinate, and acetate ions have been fully removed.
  • the preferably produced highly concentrated protein solution is then, for example, composed solely of protein, water and the necessary minimum of a selected counterion, such as chloride, citrate, succinate or acetate forming e.g. a "proteinium-chloride", “proteinium-citrate", “proteinium-succinate” or “proteinium-acetate” complex.
  • embodiments of the process of the present invention were applied to formulations of three test proteins.
  • the proteins were provided with a variety of initial buffer ions such as succinate, citrate, acetate, phosphate.
  • the exchange media was e.g. acetate, chloride, succinate.
  • Final product pools of up to 200 mg/mL protein were produced.
  • the exemplary processes were shown to consistently reduce the levels of residual initial buffer ions below the limit of detection and yielded liquid formulations with only enough exchange-medium counterions to balance the proteins' inherent charge and permit the protein to self- buffer. Protein integrity was assessed by chromatography or opalescence. In general, protein quality (as measured by monomer content) was just slightly reduced or maintained unchanged.
  • Protein 1 and Protein 2 studied were two humanized monoclonal antibodies, both of isotype IgG and subclass 1. Their average molecular weight was 150 kDa and with an isoelectric point (IP) at approximately pH 8.4 (cf. Karow A.R., Bahrenburg S., & Garidel P. (2013), Buffer capacity of biologics-from buffer salts to buffering by antibodies, Biotechnol. Prog. 29, 480-492).
  • IP isoelectric point
  • the mAbs were produced by mammalian cell culture in a CHO (Chinese hamster ovary) cell line (see Bergemann K., Eckermann C, Garidel P., Grammatikos S., Jacobi A., Kaufmann H., Kempken R., & Pisch-Heberle S. (2007), Production and Downstream Processing, in: Handbook of Therapeutic Antibodies pp. 199-237, Wiley-VCH Verlag GmbH; Jacobi A., Enenkel B., Garidel P., Eckermann C, Knappenberger M., Presser I. & Kaufmann H. (2014), Process Development and Manufacturing of Therapeutic Antibodies, in: Handbook of Therapeutic Antibodies pp.
  • Protein 3 studied was a nanobody (see e.g. Gibbs W.W. (2005), Nanobodies, Sci Am. Aug, 293 (2): 78-83), a trimer with an average molecular weight of 40.7 kDa and an isoelectric point at approximately pH 8.4 (theoretical) / pH 7.5 (experimental).
  • the nanobody was produced via E. coli microbial fermentation and is processed and purified accordingly (cf. Arbabi-Ghahroudi M., Tanha J., MacKenzie R. (2005), Prokaryotic expression of antibodies, Cancer Metastasis Rev.
  • Protein 4 is a FC fusion protein, the amino sequence is indicated in example 1 1.
  • Protein 5 is 100% identical to the published sequence of Rituximab, the heavy chain and light chain is indicated in example 12. Both sequences are listed as SEQ ID No. 3 ("Artificial Sequence", "FC fusion protein”), SEQ ID No.
  • Succinic acid, trichloroacetic acid, trisodium citrate dihydrate, acetic acid, and sodium acetate were purchased from Merck KGaA; citrate acid monohydrate from Jungbunzlauer Ladenburg GmbH; disodium succinate hexahydrate and monosodium phosphate from Dr. Paul Lohmann GmbH; disodium phosphate from Chemische Fabrik Budenheim KG; sucrose from Sudzucker AG; and sodium chloride from Akzo Nobel.
  • pH The room temperature pH of the protein solutions at each stage was assessed using a pH meter with coupled pH-electrode (Mettler Toledo SevenGo, Columbus, OH, USA). Before each pH measurement, it was carried out a two-point calibration with calibration solutions at pH 4 and pH 7 (Mettler Toledo SevenGo, Columbus, OH, USA).
  • Osmolality Osmolality was determined using a freezing-point osmometer (Osmomat 030, Gonotec, Berlin, Germany).
  • Protein integrity The investigation of protein quality focused on particle formation, as indicated by visual inspection, opalescence, and high-performance size- exclusion liquid chromatography (HP-SEC) (Garidel et al. 2010, loc.cit; den Engelsman J., Garidel P., Smulders R., Koll H., Smith B., Bassarab S., Seidl A., Hainzl O., & Jiskoot W. (201 1 ), Strategies for the Assessment of Protein Aggregates in Pharmaceutical Biotech Product Development, Pharm Res 28, 920- 933).
  • Visual inspection Visual inspection was performed according to the current Pharmacopeia.
  • Opalescence may indicate protein particle formation (Sukumar M., Doyle B.L., Combs J.L., & Pekar A.H. (2004), Opalescent appearance of an lgG1 antibody at high concentrations and its relationship to noncovalent association, Pharmaceutical Research 21 , 1087-1093). An increase in opalescence is mostly linked to an increase in protein aggregation or particle concentration. Opalescence is measured in formazine nephelometric units (FNU) via photometry of 90°- scattered light at 400-600 nm. The photometer (2100AN Laboratory Turbidimeter, Hach, Loveland, CO, USA) was initially calibrated with standards for 20 and 100 FNU, according to the European Pharmacopoeia (2013).
  • FNU formazine nephelometric units
  • HP-SEC High-performance size-exclusion liquid chromatography
  • Monomer content and levels of protein aggregates were determined via size-exclusion chromatography (Acquity HCIass and TUV detector, both from Waters Corporation, Milford, MA, USA; columns were Waters Acquity UPLC 4.6 mm x 300 mm analytical column, also from Waters Corporation, Milford, MA, USA).
  • the mobile phase was 200 mM L-arginine, 120 mM ammonium sulfate, and 10% isopropyl alcohol adjusted with 85% phosphoric acid to pH 7.3.
  • the HP- SEC was performed at room temperature.
  • SCX Strong cation exchange chromatography
  • IEC Ionic exchange chromatography Analytical strong cation exchange chromatography (SCX) was used for separation and quantification of charge variants of protein 3 8prot 3).
  • a MabPac® SCX-10 column (4 x 250 mm, 10 ⁇ , Thermo Scientific 074625) was used on an Alliance HPLC-System (Waters) coupled with a UV- detector. The column was tempered to 35°C. The separated peaks were detected at an absorbance of 280 nm. Elution was caused by a gradient from 100 % to 64 % buffer A (10 mM Na 2 HP0 4 with pH 7.0) in 22 minutes with a constant flow of 1 ml_ * min "1 .
  • the column regeneration was caused by 100 % buffer B (10 mM Na 2 HP0 , 1 M NaCI, pH 7) for 4 minutes at a constant flow of 1 ml_ * min "1 .
  • Protein 3 was diluted with water to a concentration of 0.2 mg * ml_ "1 .
  • 40 ⁇ _ of the diluted protein was injected.
  • Buffer A contained 10 mM Na2HP0 4 with pH 7.0
  • Carboxylic acid analytics (succinate, citrate, acetate): Residual excipient was determined by high pressure liquid chromatographic analysis (Akta Micro, GE Healthcare, Little Chalfont, UK; Acclaim Organic Acid Column, 5 ⁇ 4.0 x 250 mm, Thermo Fischer, Waltham, MA, USA). The mobile phase was 100 mM sodium sulfate adjusted to pH 2.6 with 99% methane sulfonic acid. The column temperature was set to 30°C by a column heater. Injection volume was 10 ⁇ _, and an isocratic elution with a flow rate of 0.6 mL/min was applied. Carboxyl group concentrations were measured by UV-Vis detector tuned to 210 nm.
  • the mAb was first precipitated with TCA (trichloro acetic acid) (10%) in a ratio of 3:1 (sample/TCA, V/V). A calibration curve was generated and used to determine the residual excipient levels.
  • the protein precipitation increases the solution's excluded volume, and this must be considered when calculating excipient concentrations. When the protein concentration is very high, these deviations can run as 15% (200 mg/mL).
  • Chloride and phosphate Ion-specific cuvette kits (LCK 31 1 for chloride, LCK 350 for phosphate, Hach, Salford, UK) were used to determine chloride and phosphate concentrations.
  • the cuvettes contain pre-dosed reagents with defined concentrations.
  • Sample preparation included a TCA protein precipitation identical to that employed in the carboxylic acid analysis, and for the same reason: proteins would precipitate with reagents in the cuvettes and would interfere photometric evaluation.
  • the LCK349 test for phosphate determines ion concentration in the 2-20 mg/L range.
  • 0.4 mL of sample is added to the cuvette, followed by 0.5 ml_ of the Solution B provided with the kit.
  • the cuvette is then sealed with a DosiCap C.
  • the phospate LOQ 0.05 mg/L, according to the present invention. After shaking and a hold time of 10 minutes, the samples were analysed with by spectral photometer (DR 3900, Hach, Salford, UK).
  • step (b) DF1 diafiltration against a buffer medium (medium B of high ionic strength and therefore high conductivity);
  • step (c) DF2, diafiltration against water (medium C of low ionic strength and therefore low conductivity);
  • step (d) UF2, ultrafiltration to concentrate the protein to the desired value.
  • a second embodiment was tested to exchange the starting buffer and concentrate the protein solution.
  • a centrifugal filtration system (regenerated cellulose, Amicon Ultra 15 ml_ Centrifugal Filter, Merck Millipore, Billerica, MA, USA) was used for a Protein 3 nanobody. The process conditions were similar to those used in the UF/DF process for the Protein 1 and Protein 2 mAbs.
  • Figure 1 A a schematic representation of a diafiltration (DF) step
  • Figure 1 B a schematic representation of an ultrafiltration (UF) step
  • FIG. 2 a schematic representation of an ultrafiltration/diafiltration (UF/DF) process according to prior art
  • FIG. 3 a schematic representation of an exemplary embodiment of the ultrafiltration/diafiltration (UF/DF) process according to the present invention
  • FIG 4A a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 1 (Protl ) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a succinate-acetate exchange;
  • Figure 4B a diagram wherein conductivity, opalescence and monomer content during the UF/DF-process of protein 1 (Protl ) (y axis) of Figure 4A are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a succinate-acetate exchange;
  • FIG. 5A a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 1 (Protl ) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a succinate-acetate exchange;
  • Figure 5B a diagram wherein conductivity, opalescence and monomer content during the UF/DF-process of protein 1 (Protl ) (y axis) of Figure 5A are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a succinate-acetate exchange;
  • FIG. 6A a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 1 (Protl ) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a citrate-chloride exchange;
  • Figure 6B a diagram wherein conductivity, opalescence and monomer content during the UF/DF-process of protein 1 (Protl ) (y axis) of Figure 6A are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a citrate-chloride exchange;
  • FIG. 7A a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 1 (Protl ) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a citrate-acetate exchange;
  • Figure 7B a diagram wherein conductivity, opalescence and monomer content during the UF/DF-process of protein 1 (Protl ) (y axis) of Figure 7A are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a citrate-acetate exchange;
  • FIG 8 a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 1 (Protl ) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a succinate-chloride exchange;
  • FIG. 9 a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 1 (Protl ) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in an acetate-chloride exchange;
  • FIG 10A a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 2 (Prot2) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a phosphate-succinate exchange;
  • Figure 10B a diagram wherein conductivity, opalescence and monomer content during the UF/DF-process of protein 2 (Prot2) (y axis) of Figure 10A are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a phosphate-succinate exchange;
  • Figure 1 1 a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 2 (Prot2) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a phosphate-citrate exchange;
  • UF/DF ultrafiltration/ diafiltration
  • FIG 12 a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 2 (Prot2) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a phosphate-chloride exchange;
  • FIG. 13A a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 2 (Prot2) (y axis) are plotted against the pH value in each process step (step (a) to step (d)) (x axis) in a succinate-chloride exchange;
  • Figure 13B a diagram wherein monomer content and IEC (ion-exchange chromatography) main peak during the UF/DF-process of protein 2
  • FIG 14 a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipient and protein concentrations during the UF/DF-process of protein 4 (Prot4) (y axis) are indicated in each process step (step (a) to step (d)) (x axis) in a phosphate-chloride exchange; and Figure 15 a diagram of an exemplary embodiment of the ultrafiltration/ diafiltration (UF/DF) process according to the present invention: the excipients and protein concentrations during the UF/DF-process of protein 5 (Prot5) (y axis) are indicated in each process step (step (a) to step (d)) (x axis) in an acetate/succinate/citrate-chloride exchange.
  • Figures 1A, 1 B, 2 and 3 have been already described. The examples according to the present invention are explained in connection with the Figures 4A to 15 in the following:
  • the water used is MilliQ® water.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial succinate buffer ions with low levels of acetate.
  • a "proteinium-acetate” formulation is generated, with acetate as counterion.
  • the used biomolecule (designated as “Protl " hereafter) was a monoclonal antibody comprising this heavy chain (amino acid single letter code, N to C- terminus):
  • SEQ ID NO. 1 (Artificial Sequence”, “monoclonal antibody, heavy chain”) and SEQ ID NO. 2 (“Artificial Sequence”, “monoclonal antibody, light chain”) in the accompanying sequence listing.
  • SEQ ID NO. 1 (Artificial Sequence”, “monoclonal antibody, heavy chain)
  • SEQ ID NO. 2 (Artificial Sequence”, “monoclonal antibody, light chain”)
  • the starting solution is 10 mg/ml_ Protein 1 mAb in ultrapure water which contains 25 mM sodium succinate and 125 mM sodium chloride at pH 6.5.
  • Figure 4A shows the results of the succinate-acetate exchange according to example 1.
  • the UF/DF-process including a diafiltration step with 4 cycles of 500 mM sodium acetate pH 5.0 (DF1) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) (pH 6) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (acetate) of DF1.
  • type 1 e.g. MilliQ® water of Merck Millipore
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: inital, end of UF1 , cycle #1 / #2 / #3 / #4 of DF1 , cycle #2 / #4 / #6 of DF2 and the final product (Prod Pool) at the end of UF2.
  • Figure 4B shows the results of the conductivity, opalescence and monomer content of the succinate-acetate exchange of example 1.
  • the initial, measured chloride concentration is 125 mM and succinate concentration is 25 mM.
  • UF1 concentrates the protein to -40 mg/ml_.
  • DF1 consists of four cycles of diafiltration against 500 mM sodium acetate at pH 5.0 to reduce succinate levels to 0.5 mM and chloride concentration below the detection limit. Increasing the number of diafiltration cycles would further reduce succinate concentration, but at the expense of process time and potential protein stress.
  • UF2 follows, concentrating Protein 1 to ⁇ 150 mg/ml_. With this volume reduction, acetate concentration increases to -26 mM. The resulting acetate/protein ratio is -26:1 , very close to the level for the 40 mg/ml_ Protein 1 solution at the end of DF2: this is the proportion of acetate counterions required to maintain the system's charge neutrality. The observed difference between 30:1 (after DF2) and 26:1 (after UF2) may be attributed to minor errors in measuring anion and/or protein concentrations. At this stage, the product pool (final product) is 150 mg/ml_ Protein 1 mAb at pH 5.9 with -20 mM acetate serving as counterions.
  • the pH of the product pool is determined and maintained by the protein's own self-buffering capacity and the counterion (Karow et al. 2013, loc.cit).
  • Product quality is monitored via opalescence and HP-SEC monomer content.
  • Conductivity is used for process control. As expected, after DF2, conductivity decreases from 15 millisiemens per centimetre (mS-crrf 1 ) to close to 1 mS-cm "1 .
  • mS-crrf 1 millisiemens per centimetre
  • opalescence increases from 8 FNU at 10 mg/ml_ to more than 20 FNU at 40 mg/ml_ in the presence of succinate. Exchanging succinate for acetate in DF1 reduces opalescence to -17 FNU.
  • Figure 4B illustrates the good product quality with regard to the high degree of monomer content throughout the steps of the process.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial succinate buffer ions with low levels of acetate, wherein the pH in the DF1 step was changed.
  • UF2 Product Pool: 138 mg-ml "1 Protl / 20 mM Acetate / water / pH 6.4.
  • Figure 5A shows the results of the succinate-acetate exchange at higher pH according to example 2.
  • the excipient and protein concentrations during the UF/DF-process of protein 1 are entered, the UF/DF-process including a diafiltration step with 4 cycles of 500 mM sodium acetate pH 6.0 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g.
  • MilliQ® water of Merck Millipore pH 6
  • DF2 MilliQ® water of Merck Millipore
  • the concentration of the anion is dependent on the amount of the positive net charge of the protein, which is mainly influenced by the pH and the concentration of the protein.
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: inital, end of UF1 , cycle #1 / #3 / #4 of DF1 , cycle #2 / #6 of DF2 and the final product (ProdPool) at the end of UF2.
  • Figure 5B shows the results of the conductivity, opalescence and monomer content of the succinate-acetate exchange of example 2.
  • Figure 5B also illustrates the good product quality with regard to the high degree of monomer content throughout the steps of the process.
  • UF2 Product Pool: 144 mg-ml "1 Protl / 20 mM Chloride / water / pH 5.8.
  • Figure 6A shows the results of the citrate-chloride exchange according to example 3.
  • the UF/DF-process including a diafiltration step with 4 cycles of 500 mM NaCI pH 6.0 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) (pH 6) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (chloride) of DF1.
  • type 1 e.g. MilliQ® water of Merck Millipore
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: inital, end of UF1 , end of DF1 , end of DF2 and the final product (ProdPool) at the end of UF2.
  • Figure 6B shows the results of the conductivity, opalescence and monomer content of the citrate-chloride exchange of example 3.
  • citrate was cleared and exchanged for chloride (Figure 6A).
  • the initial protein solution is composed of 10 mg/ml_ Protein 1 mAb with 48 mM sodium citrate without additional salt at pH 6.5 ( Figure 6A).
  • UF1 concentrated the protein to 40 mg/ml_ mAb
  • DF1 is run against 500 mM sodium chloride at pH 6.0. After 4 diafiltration cycles, citrate concentration is reduced to 2 mM.
  • the final product pool is 144 mg/ml_ Protein 1 at pH 5.8 with 20 mM chloride anions as counterions ( Figure 6A), at a chloride/protein ratio of 21 :1 to 26:1.
  • the indices of product quality show a small (0.4%) decrease in monomer content over the course of the four steps. Such decreases are not unusual when proteins are concentrated to the 100 mg/ml_ level. The degree depends on the target protein concentration, the protein's sensitivity to the shear stresses of UF/DF, the buffer, and process conditions (e.g. membrane material, transmembrane pressure, and flux).
  • Figure 6B also illustrates the good product quality with regard to the high degree of monomer content throughout the steps of the process.
  • FIG. 7A shows the results of the citrate-acetate exchange according to example 4.
  • the UF/DF-process including a diafiltration step with 4 cycles of 500 mM sodium acetate pH 6.0 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) (pH 6) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (acetate) of DF1.
  • type 1 e.g. MilliQ® water of Merck Millipore
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: inital, end of UF1 , end of DF2, and the final product (ProdPool) at the end of UF2.
  • Figure 7B shows the results of the conductivity, opalescence and monomer content of the citrate-acetate exchange of example 4.
  • the initial citrate buffer is exchanged for acetate and cleared ( Figure 7A).
  • the initial solution is 10 mg/ml_ Protein 1 with 48 mM sodium citrate at pH 6.1 .
  • DF1 40 mg/ml_ protein solution is diafiltered against 500 mM sodium acetate at pH 6.0. The citrate is easily removed, falling below the detection limit after six DF2 cycles. And after UF2, the final product pool is 160 mg/ml_ Protein 1 at pH 6.4 with 23 mM acetate counterions (for an acetate/protein ratio of about 22: 1 ).
  • Figure 7B also illustrates the good product quality with regard to the high degree of monomer content throughout the steps of the process.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial succinate buffer ions with chloride.
  • DF1 4 cycles with 500 mM NaCI / water / pH 6.2; DF2: 6 cycles with water;
  • Figure 8 shows the results of the succinate-chloride exchange according to example 5.
  • the excipient and protein concentrations during the UF/DF-process of protein 1 (Protl ) are entered, the UF/DF-process including a diafiltration step with 4 cycles of 500 mM NaCI pH 6.2 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) (pH 6) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (chloride) of DF1.
  • the concentration of the anion is dependent on the amount of the positive net charge of the protein, which is mainly influenced by the pH and the concentration of the protein.
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: inital, end of UF1 , end of DF1 , end of DF2 and the final product (ProdPool) at the end of UF2.
  • example 6 an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial accetate buffer ions with chloride.
  • the detailed conditions of example 6 were as follows:
  • UF2 Product Pool: 157 mg-ml "1 Protl / 23 mM Chloride / water / pH 5.7.
  • Figure 9 shows the results of the acetate-chloride exchange according to example 6.
  • the excipient and protein concentrations during the UF/DF-process of protein 1 are entered, the UF/DF-process including a diafiltration step with 4 cycles of 500 mM NaCI pH 6.0 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g.
  • MilliQ® water of Merck Millipore pH 6
  • DF2 MilliQ® water of Merck Millipore
  • the concentration of the anion is dependent on the amount of the positive net charge of the protein, which is mainly influenced by the pH and the concentration of the protein.
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: inital, end of UF1 , end of DF1 , end of DF2, and the final product (ProdPool) at the end of UF2.
  • the exchange of chloride for succinate ( Figure 8) and chloride for acetate ( Figure 9) were evaluated.
  • DF1 is run for four cycles against 500 mM sodium chloride, followed by six DF2 cycles against pure water.
  • the initial buffer ion is fully removed and the product pool is 157 mg/ml_ Protein 1 , with a chloride/Protein 1 ratio of about 20:1.
  • the indices of product quality were within expected ranges and were not impaired by the process (data not shown).
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial phosphate buffer ions with succinate.
  • a second antibody, the lgG1 mAb Protein 2 was tested to investigate double- diafiltration UF/DF performance with another protein ( Figures 10A and 10 B).
  • FIG. 10A shows the results of the phosphate-succinate exchange according to example 7.
  • the UF/DF-process including a diafiltration step with 4 cycles of 500 mM sodium succinate pH 5.7 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) (pH 6) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (succinate) of DF1.
  • type 1 e.g. MilliQ® water of Merck Millipore
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: initial, end of UF1 , cycle #1 / #2 / #3 / #4 of DF1 , cycle #1 / #2 / #3 / #4 / #5 / #6 of DF2 and the final product (ProdPool) at the end of UF2.
  • Figure 10B shows the results of the conductivity, opalescence and monomer content of the phosphate-succinate exchange of example 7.
  • Example 7 began with an initial solution of 20 mg/ml_ Protein 2 in 13 mM sodium phosphate and 146 mM sucrose at pH 7.3 (Wang W. (1999) Instability, stabilization, and formulation of liquid protein pharmaceuticals. International Journal of Pharmaceutics 185, 129-188.).
  • UF1 concentrated the protein to more than 30 mg/ml_. DF1 , run against 500 mM sodium succinate at pH 5.7, then completely removed the phosphate. After DF2 against water and UF2, the product pool was 89 mg/mL Protein 2 with 4 mM succinate and a succinate/protein ratio below 10: 1 (Note that under these pH conditions, succinate has a charge of -2.).
  • Figure 10B also illustrates the good product quality with regard to the high degree of monomer content throughout the steps of the process.
  • UF2 (Product Pool): 64 mg-ml "1 Prot2 ⁇ .5 mM Citrate / water / pH 7.0.
  • Figure 1 1 shows the results of the phosphate-citrate exchange according to example 8.
  • the UF/DF-process including a diafiltration step with 4 cycles of 500 mM sodium citrate pH 6.0 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) (pH 7) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (citrate) of DF1.
  • type 1 e.g. MilliQ® water of Merck Millipore
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: initial, end of UF1 , cycle #1 / #2 / #3 / #4 of DF1 , cycle #1 / #2 / #3 / #4 / #5 / #6 of DF2 and the final product (ProdPool) at the end of UF2.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial phosphate buffer ions with chloride.
  • Figure 12 shows the results of the phosphate-chloride exchange according to example 9.
  • the excipient and protein concentrations during the UF/DF-process of protein 2 (Prot2) are entered, the UF/DF-process including a diafiltration step with 4 cycles of 500 mM NaCI pH 7.0 (DF1 ) followed by 6 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) (pH 7) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (chloride) of DF1.
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: initial, end of UF1 , cycle #1 / #2 / #3 / #4 of DF1 , cycle #1 / #2 / #3 / #4 / #5 / #6 of DF2 and the final product (ProdPool) at the end of UF2.
  • Phosphate can be completely removed by exchange with either citrate or chloride ( Figures 11 and 12).
  • the initial solution of Protein 2 and phosphate was diafiltered against citrate. Viscosity increased, throughput fell, and the concentration of the final product pool was just 64 mg/ml_ Protein 2.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial succinate buffer ions with chloride.
  • FIG. 13A shows the results of the succinate-chloride exchange according to example 10.
  • the excipient and protein concentrations during the UF/DF-process of protein 3 are entered, the UF/DF-process including a diafiltration step with 8 cycles of 200 mM NaCI pH 4.5 (DF1 ) followed by 5 cycles of diafiltration with ultrapure water of type 1 (e.g.
  • MilliQ® water of Merck Millipore pH 4.5
  • DF2 MilliQ® water of Merck Millipore
  • the x axis coordinates the process steps and the corresponding pH.
  • the respective points on the x axis are: initial, end of UF1 , end of DF1 , end of DF2 and the final product (ProdPool) at the end of UF2.
  • Figure 13B shows the results of the monomer content and IEC main peak of the succinate-chloride exchange of example 10.
  • example 10 it was tested the UF/DF process at the small scale, using Amicon ultra centrifugal filter units to condition and concentrate Protein 3, the nanobody. As example 10, it was assessed DF1 replacement of succinate with chloride (Figure 13A).
  • the initial protein solution is 8 mg/mL Protein 3 in 25 mM succinate under acidic conditions (pH 4.4).
  • the UF1 step increased concentration to more than 45 mg/mL Protein 3. Because Protein 3 showed solubility problems at high ionic strength, DF1 was run against 200 mM sodium chloride for 8 cycles; this was sufficient to fully remove succinate. Five DF2 cycles against pure water reduced the chloride content to 13 mM. UF2 brought the concentration of the final product pool to 125 mg/mL Protein 3 in -30 mM chloride, with a chloride/protein ratio between 10:1 and 14:1. Under these process conditions the main peak of the ionic exchange peak is unchanged and the amount of aggregates as measured by high performance size exclusion chromatography (HP-SEC) is just reduced by 0.5%-0.8%, which is considered highly acceptable for a nanobody (Figure 13B).
  • HP-SEC high performance size exclusion chromatography
  • Figure 5B also illustrates the good product quality with regard to the high degree of monomer content throughout the steps of the process.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial phosphate buffer ions with chloride.
  • the used biomolecule (designated as "Prot4" hereafter) was a Fc fusion protein.
  • the amino acid sequence of the FC fusion protein was as follows:
  • SEQ ID NO. 3 (Artificial Sequence", “FC fusion protein”) in the accompanying sequence listing.
  • UF2 (Product Pool): 212 mg-ml "1 Prot4 / water / pH 7.2.
  • Figure 14 shows the results of the phosphate-chloride exchange according to example 1 1.
  • the UF/DF-process including a diafiltration step with 4 cycles of 500 mM NaCI pH 7.0 (DF1 ) followed by 8 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) including 0.002 wt% NaCI (pH 7) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (chloride) of DF1.
  • type 1 e.g. MilliQ® water of Merck Millipore
  • the x axis indicates the process steps.
  • the respective points on the x axis are: initial, end of UF1 , cycle #1 / #2 / #3 / #4 of DF1 , cycle #1 / #2 / #3 / #4 / #5 / #6 / #7 / #8 of DF2, sampling point #1 / #2 / #3 / #4 / #5 of UF2 and the final product (ProdPool) at the end of UF2.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial acetate, succinate and citrate buffer ions with chloride.
  • the used biomolecule (designated as "Prot5" hereafter) had a sequence which was 100% identical to the published sequence of Rituximab comprising this heavy chain (amino acid single letter code, N to C-terminus):
  • SSPVTKSFNR GEC The sequences are listed as SEQ ID NO. 4 ("Artificial Sequence”, “Rituximab HC”) and SEQ ID NO. 5 ("Artificial Sequence”, “Rituximab LC”) in the accompanying sequence listing.
  • Figure 15 shows the results of the acetate/succinate/citrate-chloride exchange according to example 12.
  • the excipient concentrations the protein concentrations and the monomer content during the UF/DF-process of protein 5 (Prot5) are entered, the UF/DF-process including a diafiltration step with 4 cycles of 500 mM NaCI pH 7.0 (DF1 ) followed by 8 cycles of diafiltration with ultrapure water of type 1 (e.g. MilliQ® water of Merck Millipore) including 0.002 wt% NaCI (pH 7) (DF2) to provide an entire exchange of the excipients from the initial solution towards the anion component (chloride) of DF1.
  • type 1 e.g. MilliQ® water of Merck Millipore
  • the x axis indicates the process steps.
  • the respective points on the x axis are: initial, end of UF1 , cycle #1 / #2 / #3 / #4 of DF1 , cycle #1 / #2 / #3 / #4 / #5 / #6 / #7 / #8 of DF2, and the final product (ProdPool) at the end of UF2.
  • the three carboxylic acids acetate, succinate and citrate can be completely removed by exchange with chloride.
  • UF2 the chloride ions get concentrated in the same way the protein was concentrated.
  • the concentration of the anions acetate, succinate and citrate remains under the limit of quantification (LOQ). Due to the acid pH of 4.9 at the end of UF2 the amount of the counterions chloride showed a high level of 40 mM.
  • the presented examples show that the process according to the present invention can be used for antibodies as well as non-antibody formats. It allows to conditioning clearly defined formulations and by spiking additional excipients specific, well defined formulations can be generated.
  • the process was repeated 3 times to verify the robustness of the process. That is, at first, the process of the present invention including steps (a) to (d) comprising the order UF1/DF1/DF2/UF2 was performed and the resulting (first) biomolecule formulation investigated. Then, the same process using the same starting material and the same conditions was repeated and the resulting (second) biomolecule formulation investigated. Finally, the same process using the same starting material and the same conditions was again repeated and the resulting (third) biomolecule formulation investigated.
  • an embodiment of the 4-step UF/DF process according to the present invention was applied to concentrate a protein and replace initial accetate buffer ions with chloride.
  • the same procedure as already described in example 6 was performed but the detailed conditions were selected to be as follows:
  • the invention comprises aspects which are disclosed in the sentences below:
  • a process for the preparation of a highly concentrated liquid formulation containing biomolecules comprising the steps of
  • step (d) a second ultrafiltration UF2; whereby an aqueous solution of one or more salts as liquid medium B is used for step (b) and water or an aqueous solution of one or more salts as liquid medium C is used for step (c), whereby the salts used for step (b) are the same or different from the salts used for step (c), the liquid medium B has an ionic strength which is higher than the ionic strength of the liquid medium C.
  • the liquid medium B has a high ionic strength indicated in form of a concentration which is in the range of from about 20 mM up to the limit of solubility of the salt, particularly preferred from about 100 mM to 1000 mM, more preferred about 150 mM to 750 mM, most preferred from about 200 mM to 500 mM
  • the liquid medium C has a low ionic strength indicated in form of a concentration which is in the range of from about 0 mM to 150 mM, particularly preferred from about 0 mM to 100, more preferred about 0 mM to 75 mM, most preferred from about 0 mM to 50 mM.
  • the liquid medium B has an ionic strength which is higher than the ionic strength of the liquid medium C so that the difference between the ionic strength of the liquid medium B and the ionic strength of the liquid medium C indicated in form of a concentration is at least about 100 mM, more preferred at least about 200 mM, most preferred at least about 500 mM.
  • the liquid biomolecule formulation used in step (a) contains a liquid medium A which is an aqueous solution and contains one or more excipients, the liquid medium A is exchanged with liquid medium C by means of liquid medium B in steps (b) and (c), whereby the liquid biomolecule formulation obtained in step (c) and (d) has a reduced content of said excipient(s).
  • the excipients are selected from a group consisting of
  • excipients charged or neutral in aqueous solution preferably the excipients being selected from the group consisting of additives used in the preparation or processing of biomolecules; unwanted substances or compounds such as impurities contained in the starting liquid biomolecule formulation; undesired side-products formed during the manufacturing process of the biomolecule; decomposition or degradation products of starting, intermediate or end products formed during the production of the biomolecule; particularly preferred cell components or debris, degradation products of bacteria such as endotoxines, DNA, RNA, undesired lipids, HCP (Host cell proteins), lipopolysaccharides (LPS) or parts thereof; sugars; detergents such as positively charged, negatively charged and also non-ionic species; any kind of negatively or positively charged ions, preferably resulting from salts.
  • the excipients being selected from the group consisting of additives used in the preparation or processing of biomolecules; unwanted substances or compounds such as impurities contained in the starting liquid biomolecule formulation; undesired side-products formed during the manufacturing process of the
  • the salts are selected from organic salts and/or inorganic salts.
  • the inorganic salt is selected from the group consisting of alkali salts or alkaline earth salts of sulfates, nitrates, phosphates, carbonates, halogenides, borates, silkates and the like
  • the inorganic salt is selected from the group of pharmaceutically acceptable inorganic salts, preferably sodium salts such as sodium halides, particularly preferred sodium chloride, sodium sulfate, sodium borate; calcium salts such as calcium halides, particularly preferred calcium chloride, calcium sulfate, calcium borate; magnesium salts such as magnesium halides, particularly preferred magnesium chloride, magnesium sulfate, magnesium borate, and combinations thereof,
  • the inorganic salt is sodium chloride.
  • liquid medium B comprises sodium chloride in a concentration from about 150 to about 900 mM, increasingly preferred from about 200 to about 700 mM, from about 400 to about 600 mM, and from about 450 to about 550 mM.
  • the salt is an organic and/or inorganic buffer salt.
  • the buffer salt is the basis of a buffer, preferably biological buffer, selected from the group consisting of N-(2-acetamido)-aminoethanesulfonic acid (ACES) and salts thereof, acetic acid and salts thereof, aconitic acid and salts thereof, adipic acid and salts thereof, ascorbic acid and salts thereof, A/-(2-Acetamido)-iminodiacetic acid (ADA) and salts thereof, ammonia and salts thereof, ammonium chloride, 2-amino-2-methyl-1 -propanol (AMP), 2- amino-2-methyl-1 ,3-propanediol, ammediol (AMPD), A/-(1 ,1 -Dimethyl-2- hydroxyethyl)-3-amino-2-hydroxypropanesulfonic acid (AMPSO) and salts thereof, A/,A/-bis-(2-hydroxyethyl)-2-aminoethanesulfonic acid (BES
  • the biological buffer is an amino acid in an aqueous solution, the amino acid being selected from the group consisting of alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine;
  • particularly preferred biological buffers are selected from the group consisting of phosphoric acid and salts thereof, citric acid and salts thereof, tris, succinic acid and salts thereof, malic acid and salts thereof, tartaric and salts thereof, acetic acid and salts thereof, lactic acid and salts thereof, aconitic acid and salts thereof, ascorbic acid and salts thereof, glutamic acid and salts thereof, ammoniumchloride, triethanolamine, alanine, arginine, glutamine, glycine, histidine, lysine, and proline.
  • the liquid medium C consists or essentially consist of water.
  • the biomolecule and the excipient(s) to be removed from the liquid biomolecule formulation have opposite charges
  • the biomolecule is positively charged and the excipient(s) to be removed by the process are negatively charged excipient(s),
  • biomolecule is a positively charged protein and the negatively charged excipient(s) are anions.
  • x 2 to 10
  • x 2 to 8
  • x 2 to 6
  • y 2 to 10
  • y 2 to 8
  • y 2 to 6
  • the ultrafiltration UF1 of step (a) is used to concentrate the liquid biomolecule formulation, preferably up to about 10%-70%, more preferably about 15%-60%, most preferably about 25%-50% compared with the initial concentration of the liquid biomolecule formulation.
  • the ultrafiltration UF2 of step (d) is used to concentrate the liquid biomolecule formulation to the desired value.
  • step (b) and step (c) follow directly one after the other whereby no intermediate process step is performed in between,
  • step (a) and step (b) follow directly one after the other whereby no intermediate process step is performed in between, and preferably also step (c) and step (d) directly follow one after the other whereby no intermediate process step is performed in between.
  • biomolecules are selected from the group consisting of
  • lipds such as phospholipids, glycolipids, sterols; vitamins; hormones; neurotransmitter;
  • Monomers preferably amino acids, nucleotides, monosaccharides; biopolymers, preferably proteins or peptides; nucleic acids such as DNA, RNA; oligosaccharides, polysaccharides such as glycogen, starch, chitin, cellulose, fructane, dextrane;
  • proteins or peptides particularly preferred are proteins or peptides, nucleic acids, oligosaccharides, and polysaccharides;
  • the process steps (a) to (d) are performed using a tangential flow filtration (TFF) system or a centrifugal filtration system.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Analytical Chemistry (AREA)
  • Water Supply & Treatment (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Inorganic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne un procédé en plusieurs étapes amélioré pour la préparation d'une formulation liquide hautement concentrée contenant des biomolécules comprenant les étapes consistant à (a) une première ultrafiltration UF1; (b) une première diafiltration DF1; (c) une seconde diafiltration DF2; et (d) une seconde ultrafiltration UF2; une solution aqueuse d'un ou de plusieurs sels, en tant que milieu liquide B, étant utilisée pour l'étape (b) et de l'eau ou une solution aqueuse d'un ou de plusieurs sels, en tant que milieu liquide C, est utilisée pour l'étape (c), où un sel ou plus utilisés pour l'étape (b) étant identiques ou différents d'un ou des sels utilisés pour l'étape (c) et le milieu liquide B ayant une force ionique qui est supérieure à la force ionique du milieu liquide C. Le procédé selon la présente invention permet la préparation de formulations hautement concentrées bien définies contenant des biomolécules, en particulier des protéines, destinées à une utilisation pharmaceutique ou non pharmaceutique. Un excipient(s) indésirable(s) de la formulation de biomolécule liquide de départ, peut être réduit dans des conditions de solution, à des niveaux très faibles ou à des niveaux faibles inférieurs à la limite de détection.
EP17754327.9A 2016-08-17 2017-08-11 Procédé de préparation de formulations liquides hautement concentrées contenant des biomolécules Pending EP3500588A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16184502 2016-08-17
PCT/EP2017/070423 WO2018033482A1 (fr) 2016-08-17 2017-08-11 Procédé de préparation de formulations liquides hautement concentrées contenant des biomolécules

Publications (1)

Publication Number Publication Date
EP3500588A1 true EP3500588A1 (fr) 2019-06-26

Family

ID=56740111

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17754327.9A Pending EP3500588A1 (fr) 2016-08-17 2017-08-11 Procédé de préparation de formulations liquides hautement concentrées contenant des biomolécules

Country Status (9)

Country Link
US (1) US11572385B2 (fr)
EP (1) EP3500588A1 (fr)
JP (2) JP7114567B2 (fr)
KR (1) KR102457855B1 (fr)
CN (2) CN110198952B9 (fr)
AU (2) AU2017313268B2 (fr)
CA (1) CA3031860A1 (fr)
SG (1) SG11201901081XA (fr)
WO (1) WO2018033482A1 (fr)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2019242601A1 (en) * 2018-03-26 2020-11-12 Boehringer Ingelheim Animal Health USA Inc. Method of producing an immunogenic composition
MA52204A (fr) * 2018-04-12 2021-02-17 Amgen Inc Procédés de préparation de compositions protéiques stables
EP3790532A1 (fr) 2018-05-10 2021-03-17 Regeneron Pharmaceuticals, Inc. Formulations contenant des protéines de fusion du récepteur vegf à haute concentration
CN111096456A (zh) * 2020-02-14 2020-05-05 江西师范大学 一种荷叶中抑制晚期糖基化产物形成活性组分的制备方法
KR20240050373A (ko) 2021-09-02 2024-04-18 베링거 인겔하임 인터내셔날 게엠베하 미래의 색값 또는 대응하는 특성을 결정하는 방법 및 이를 위한 장치
WO2024008256A1 (fr) * 2022-07-05 2024-01-11 Lihme Protein Solutions Appareil amélioré pour la séparation de composés

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ199058A (en) * 1980-12-05 1985-04-30 Schreiber Foods Inc Producing cheese or a cheese base containing casein and whey proteins
SE500110C2 (sv) 1989-06-27 1994-04-18 Kabi Pharmacia Ab Sätt att rena ett protein från därtill bundna flervärda metalljoner
US5728553A (en) * 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
US5564414A (en) 1994-05-26 1996-10-15 Walker; William F. Pressurized and metered medication dose counter on removable sleeve
US6312755B1 (en) * 1999-07-16 2001-11-06 Ampc Whey treatment process for achieving high concentration of α-lactalbumin
US6693173B2 (en) 2000-12-26 2004-02-17 Alpha Therapeutic Corporation Method to remove citrate and aluminum from proteins
GB0113179D0 (en) * 2001-05-31 2001-07-25 Novartis Ag Organic compounds
US6712963B2 (en) * 2002-06-14 2004-03-30 Scilog, Llc Single-use manifold for automated, aseptic transfer of solutions in bioprocessing applications
US7241874B2 (en) 2002-06-26 2007-07-10 Zimmer Ortho Biologics, Inc. Rapid isolation of osteoinductive protein mixtures from mammalian bone tissue
ES2359576T5 (es) 2002-07-31 2020-03-03 Chiesi Farm Spa Inhalador de polvo
US7674885B2 (en) 2002-11-01 2010-03-09 Bayer Healthcare Llc Process for concentration of macromolecules
CA2554883C (fr) * 2004-01-30 2013-10-01 Zymenex A/S Production et purification d'arylsulfatase a recombinante
US20060051347A1 (en) * 2004-09-09 2006-03-09 Winter Charles M Process for concentration of antibodies and therapeutic products thereof
AU2005229674B2 (en) * 2004-11-18 2010-11-04 Kedrion Melville Inc. Low concentration solvent/detergent process of immuneglobulin with pre-treatment
CN101247897B (zh) 2005-08-24 2013-06-12 贝林格尔·英格海姆国际有限公司 雾化器
WO2007090883A1 (fr) * 2006-02-10 2007-08-16 Boehringer Ingelheim International Gmbh Formulation à libération prolongée
US20070246406A1 (en) * 2006-03-31 2007-10-25 Dibel Kevin R Tangential flow filtration apparatuses, systems, and processes for the separation of compounds
ES2294976B1 (es) 2007-11-12 2008-12-16 Grifols, S.A. "procedimiento de obtencion de albumina humana de alta eficacia para su uso en terapia de detoxificacion".
US8420081B2 (en) * 2007-11-30 2013-04-16 Abbvie, Inc. Antibody formulations and methods of making same
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
US9827384B2 (en) 2011-05-23 2017-11-28 Boehringer Ingelheim International Gmbh Nebulizer
WO2012161685A1 (fr) 2011-05-23 2012-11-29 Boehringer Ingelheim International Gmbh Nébuliseur
US10080853B2 (en) 2011-05-23 2018-09-25 Boehringer Ingelheim International Gmbh Nebulizer
JP2013043865A (ja) 2011-08-25 2013-03-04 Osaka Prefecture Univ 血中脂質降下剤及び高分子アディポネクチン産生促進剤
GB201115874D0 (en) 2011-09-14 2011-10-26 Astrazeneca Ab Inhaler
GB201122430D0 (en) 2011-12-23 2012-02-08 Xstalbio Ltd Reconstitution method for high concentration dry protein formulation
JP6345184B2 (ja) * 2012-10-30 2018-06-20 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 2段階接線流限外濾過を使用するポリペプチドの精製

Also Published As

Publication number Publication date
US11572385B2 (en) 2023-02-07
JP2022120016A (ja) 2022-08-17
AU2024202299A1 (en) 2024-05-02
US20190309016A1 (en) 2019-10-10
CA3031860A1 (fr) 2018-02-22
SG11201901081XA (en) 2019-03-28
JP7114567B2 (ja) 2022-08-08
AU2017313268B2 (en) 2024-01-18
KR102457855B1 (ko) 2022-10-25
KR20190038921A (ko) 2019-04-09
CN110198952B (zh) 2024-03-01
CN110198952B9 (zh) 2024-05-28
CN110198952A (zh) 2019-09-03
WO2018033482A1 (fr) 2018-02-22
AU2017313268A1 (en) 2019-02-07
CN118085011A (zh) 2024-05-28
JP2019535641A (ja) 2019-12-12

Similar Documents

Publication Publication Date Title
AU2017313268B2 (en) Process for the preparation of highly concentrated liquid formulations containing biomolecules
EP2997036B1 (fr) Purification de polypeptides produits par recombinaison
JP2012511531A (ja) 賦形剤不含抗体溶液を得るための方法
IL258311A (en) A method for preparing a therapeutic protein formulation and an antibody formulation produced by the method
TWI625335B (zh) 純化抗體的方法
CN111479829A (zh) 具有降低的生物活性的抗体变体和同种型
EP3060578A1 (fr) Purification d'anticorps
CN111491951B (zh) 通过疏水相互作用色谱法耗尽轻链错配的抗体变体
JP2021535739A (ja) 下流精製でのラマン分光法の使用
EP2483304B1 (fr) Réglage de filtration préalable de solutés issus de tampon pour la preparation a forte teneur en immunoglobuline
CN116348486A (zh) 在蛋白质纯化过程中降低宿主细胞蛋白质含量的方法
JP5205470B2 (ja) 免疫グロブリン凝集物
JP7295943B2 (ja) タンパク質含有溶液精製用ポリアミド媒体及びその製造方法
TW202140511A (zh) 減少來自重組蛋白生產過程中的雜質之方法
Bhiman Optimisation of ultrafiltration for human serum albumin at NBI

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190220

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20221117